Next Article in Journal
How Broadly Neutralising Antibodies Are Redefining Immunity to Influenza
Previous Article in Journal
Rat as a Predictive Model for Human Clearance and Bioavailability of Monoclonal Antibodies
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Challenges and Insights in Absolute Quantification of Recombinant Therapeutic Antibodies by Mass Spectrometry: An Introductory Review

1
Federal Institute of Material Testing and Research (BAM), 12489 Berlin, Germany
2
Leibniz-Institut für Analytische Wissenschaften—ISAS—e.V., 44139 Dortmund, Germany
*
Author to whom correspondence should be addressed.
Antibodies 2025, 14(1), 3; https://doi.org/10.3390/antib14010003
Submission received: 15 November 2024 / Revised: 21 December 2024 / Accepted: 24 December 2024 / Published: 7 January 2025

Abstract

:
This review describes mass spectrometry (MS)-based approaches for the absolute quantification of therapeutic monoclonal antibodies (mAbs), focusing on technical challenges in sample treatment and calibration. Therapeutic mAbs are crucial for treating cancer and inflammatory, infectious, and autoimmune diseases. We trace their development from hybridoma technology and the first murine mAbs in 1975 to today’s chimeric and fully human mAbs. With increasing commercial relevance, the absolute quantification of mAbs, traceable to an international standard system of units (SI units), has attracted attention from science, industry, and national metrology institutes (NMIs). Quantification of proteotypic peptides after enzymatic digestion using high-performance liquid chromatography-tandem mass spectrometry (HPLC-MS/MS) has emerged as the most viable strategy, though methods targeting intact mAbs are still being explored. We review peptide-based quantification, focusing on critical experimental steps like denaturation, reduction, alkylation, choice of digestion enzyme, and selection of signature peptides. Challenges in amino acid analysis (AAA) for quantifying pure mAbs and peptide calibrators, along with software tools for targeted MS data analysis, are also discussed. Short explanations within each chapter provide newcomers with an overview of the field’s challenges. We conclude that, despite recent progress, further efforts are needed to overcome the many technical hurdles along the quantification workflow and discuss the prospects of developing standardized protocols and certified reference materials (CRMs) for this goal. We also suggest future applications of newer technologies for absolute mAb quantification.

1. Historical Overview: Evolution of Recombinant mAbs

The development of hybridoma technology by Köhler and Milstein in 1975 provided the cornerstone for the generation of monoclonal antibodies. By fusing B lymphocytes from immunized animals with myeloma cells, stable cell lines for unlimited monoclonal antibody (mAb) production could be obtained for the first time [1]. Through their ability to bind with high selectivity and affinity to a wide range of targeting biomolecules, mAbs gained increasing attention for application in life science and medicine. The first murine mAb approved by the Food and Drug Administration (FDA) for therapeutic application in humans was Muromonab in 1986 [2]. Unfortunately, the murine nature of antibodies generated by hybridoma technology bore some disadvantages for their human therapeutic application. The immune system recognized the injected mAbs as foreign proteins, resulting in the production of antibodies against those, the so-called human anti-mouse antibody (HAMA) response [3]. Consequently, the administered antibodies were eliminated from the body, reducing the effectiveness of treatment or causing allergic reactions [4].
To overcome such limitations, the field of recombinant DNA technologies offers a wide variety of tools for structural modifications of murine mAbs to create more human-like antibodies (Figure 1) and enable large-scale production using mammalian cell lines. Initial achievements have been made by combining the variable light (VL) and heavy (VH) domains of murine mAb ① with a constant domain of human light chain (VC) and FC-region (fragment crystallizable) to generate chimeric antibodies ② with reduced but still observable immunogenicity in humans [5,6]. The first FDA-approved chimeric antibodies were Abciximab in 1994 [7] and Rituximab in 1997, a recombinant antibody produced in Chinese hamster ovary (CHO) cells [8]. Further developments enabled the reduction of murine parts in mAbs, such as complementarity-determining region (CDR) grafting, whereby it was possible to incorporate hypervariable loops from VL and VH of the antigen-binding sites (Fab) of the murine antibody into a fully human antibody scaffold [9] (Figure 1, ③). These humanized antibodies were even less immunogenic than chimeric mAbs but, in some cases, showed reduced affinity to their target antigen [10]. The first FDA-approved humanized mAb was Daclizumab in 2016, an immunosuppressive agent used to reduce renal transplant rejection in patients [11]. Since then, various methods have been developed for the humanization of therapeutic antibodies to improve their properties and make them applicable to medical treatment [12].
Another notable step in the evolution of therapeutic antibodies was the development of antibody phage display technology [13], based on the previously established phage display method by Smith et al. in 1985 [14]. For this approach, a library of antibody gene fragments from immunized or non-immunized animals as well as humans is needed. Additionally, fully synthetic libraries were established, as reviewed recently by Zhang et al., 2023 [15]. The expressed antibody fragments are presented as fusion proteins with the coat protein of M13 bacteriophage surfaces and allow selection against the targeted antigen. In contrast to traditional hybridoma technology, this in vitro selection method enables the creation of mAbs even against toxic and non-immunogenic agents. The impact of phage display on the development of therapeutic antibodies has been reviewed in detail by Frenzel et al., 2016 [16].
Figure 1. Schematic overview of therapeutic mAb structure and its evolution from murine antibodies (dark blue domains) to fully human antibodies (light blue domains) with an associated decrease in immunogenicity in humans. The constant domains of the heavy chain (CH2 and CH3) are localized in the fragment crystallizable (Fc) region. The antigen-binding Fab-fragments consist of the constant (CL) and variable (VL) parts of the light chains as well as the variable (VH) and constant domain (CH1) of the heavy chain. Adapted from [17].
Figure 1. Schematic overview of therapeutic mAb structure and its evolution from murine antibodies (dark blue domains) to fully human antibodies (light blue domains) with an associated decrease in immunogenicity in humans. The constant domains of the heavy chain (CH2 and CH3) are localized in the fragment crystallizable (Fc) region. The antigen-binding Fab-fragments consist of the constant (CL) and variable (VL) parts of the light chains as well as the variable (VH) and constant domain (CH1) of the heavy chain. Adapted from [17].
Antibodies 14 00003 g001
Further inventions in the field of structural modifications of mAbs were made by Lonberg et al. in 1994. By replacing the entire murine immunoglobulin repertoire in the mouse genome with those of humans, it was possible to create fully human antibodies [18]. Such transgenic mice produce human-like antibodies after immunization, followed by conventional hybridoma technology to obtain fully human monoclonal antibodies (Figure 1, ④). Panitumumab, the first fully human mAb isolated from a transgenic XenoMouse, was FDA-approved for therapeutic application in 2006 [19]. However, this approach is limited in cases where the immunogens are toxic or show a high degree of homology between the targeted human antigen and its murine ortholog.
Despite the mentioned limitations, the creation of chimeric, humanized or fully human antibodies was a breakthrough and led to a wave of FDA-approved antibodies of Iimmunoglobulin G antibody class 1 (IgG1) and 4 (IgG4). Many of them belong to the best-selling pharmaceuticals worldwide, like the blockbuster Adalimumab, which has effectiveness against rheumatoid arthritis and other chronic or immune-inflammatory diseases [20,21] (Table 1). Other relevant antibodies, such as Pembrolizumab and Nivolumab, are used in targeted melanoma therapy [22]. Among those common anti-cancer antibodies, modified antibody-drug conjugates (ADCs) were developed for targeting cancer antigens and delivering small, chemically linked cytotoxic agents to tumor cells. Currently, only thirteen FDA-approved ADCs for numerous cancer variants are on the market [23]. Some major limitations during clinical trials were their modification heterogeneity due to non-specific drug conjugation and dynamic in vivo change after application [24]. Therefore, it is not surprising that many quantitative methods exist for the investigation of pharmacokinetic properties of ADCs in human plasma or serum [25,26,27]. With increasing market importance, analytical methods for the quantification of conventional therapeutic mAbs are also gaining relevance in clinical and pharmaceutical analysis. These methods are crucial for ensuring product quality, safety, and efficacy, particularly as mAbs are used in life-saving therapies such as cancer treatment. Nevertheless, the complex nature of mAb structures presents distinctive challenges, and highly sensitive and specific methods are essential for the precise quantification of these biomolecules.

2. Current Strategies in MS-Based Quantification of Antibodies

Over the last years, high-performance liquid chromatography coupled with tandem mass spectrometry (HPLC-MS/MS) became further established as one of the main technologies in antibody quantification [38]. Many innovative HPLC-MS/MS-based approaches were reported, focusing on more selective, more sensitive, faster and/or simplified detection of different antibody species in human blood or serum. Most of the methods continued to quantify antibodies on a peptide level; however, studies directly measuring intact or partially digested antibodies were also increasingly presented. These methods are mainly used for the quantification of antibodies with potential matrix effects, for example, in the clinical context of pharmacokinetic parameters such as antibody metabolism (half-life) or distribution in the patient’s body [39]. Moreover, several authors reported advancements in absolute amino acids analysis for the quantification of purified antibodies. In addition to the methods mentioned above, this strategy is particularly suitable for quality control of pharmaceutical antibody formulations in industry or accurate protein quantification of reference material products. Consequently, these reference materials can be employed to develop novel MS-based protein quantification techniques. Furthermore, they are metrologically traceable, thereby enhancing the comparability and reliability of measurement outcomes on a global scale.
An overview of the mentioned quantification strategies and target applications is provided in Figure 2. Developments in peptide-based methods will be discussed in detail in the next section; “intact” strategies and amino acid-based quantification will be shortly outlined after that. The selected literature was based on reviewing the methods used in the quantification of commercially important antibodies, as listed in Table 1, followed by a systematic review of primary publications that highlight problems in antibody and protein quantification from different perspectives.

2.1. Quantification of Enzymatically Digested Antibodies

One key advantage of peptide-based quantification is the high sensitivity and selectivity that can be achieved for these analytes on commonly available triple-quadrupole (QqQ) mass spectrometers. Typically, a complex biological sample such as human blood serum containing the target antibody (or antibodies) is enzymatically digested into numerous defined peptides of varying chain lengths. The peptide mixture is chromatographically separated and contains one or more abundant peptides identified as unique to the antibody of interest. These so-called “signature peptides” originate only from the antibody sequence and are used for further MS/MS analysis. For quantification, the signals of these signature peptides are related to those of spiked analog standards in known amounts, preferably represented by stable isotope-labeled (SIL) signature peptides (Section 3.2) or co-digested signature peptides of intact SIL antibody analogs (Section 3.1). By comparing the signal from the endogenous signature peptides with the SIL peptides, the exact amount of the target antibody in the sample can be determined with high precision.
To date, no universally applicable protocol exists for peptide-based mAb quantification. Instead, individual method development and validation remain necessary for each mAb, as different sample characteristics affect digestion efficiency, peptide recovery, signal-to-noise ratios, and so on [40]. This section reviews steps in sample preparation and peptide selection frequently reported as critical for robust quantification of mAbs in biological samples.

2.1.1. Purification and Enrichment

Due to interfering matrix components (proteins, lipids, DNA), HPLC-MS/MS analysis of antibodies in human serum usually requires an enrichment step in sample preparation. This can be carried out before or after the digestion step. Enrichment prior to digestion concentrates the mAb fraction and often uses magnetic beads or solid-phase extraction (SPE) cartridges, either coupled with anti-idiotypic antibodies for CDR-specific purification [41] or protein A and G that specifically bind to the constant Fc-region of mAbs [31,36,37,42]. Precipitation with organic solvents (e.g., methanol) or inorganic salts (ammonium sulfate) is an inexpensive and simple method for isolating the overall protein fraction prior to MS analysis [43]. Enrichment of targeted peptides after digestion is very commonly performed using C18-reversed phase/cation exchange SPE cartridges or tips, a step that also removes residual salts that may form excessive adducts during electrospray ionization (ESI).
It was also noted that each sample processing step carries the risk of analyte losses. For example, a significant sample loss of approx. 40% and poor quantitative precision of ±15% was observed by Heudi et al. from SPE-based peptide cleanup of digested mAb [38]. This highlights the need to optimize SPE conditions individually for each assay and to include internal standards right from the beginning of the experiment.

2.1.2. Denaturation, Reduction, and Alkylation

Most published protocols include denaturation and reduction steps prior to digestion to unfold the tertiary structure and break intramolecular disulfide bonds of proteins. This facilitates access of the enzyme to the cleavage sites, ensuring efficient digestion. For the chemical denaturation of proteins, the use of buffers containing strong chaotropic denaturants such as urea or guanidine hydrochloride [44,45] in final concentrations > 6 M and ionic detergents like sodium dodecyl sulfate (SDS) and sodium deoxycholate (DOC) [44,45] was reported. Proc et al. demonstrated that the use of guanidine hydrochloride consistently yielded lower digestion efficiency than urea, SDS or DOC-based denaturing protocols [46]. Additionally, guanidine hydrochloride can inhibit trypsin activity even at low concentrations, rendering a buffer exchange step necessary. High urea concentrations can also influence digestion efficiency, but sample dilution to a urea concentration < 1 M was sufficient for complete trypsin digestion [38]. As urea is heat-sensitive, it can degrade in different buffers into ammonium cyanate, which binds to free amines by carbamylation reactions, resulting in structure modifications of proteins [47]. However, if SDS remained in the buffer solution during trypsin digestion, the proteolytic enzyme denatured and subsequent MS analysis was affected by ionization suppression at levels as low as 0.01% [48,49].
Several MS-compatible detergents were made commercially available, such as ProteaseMax (Promega GmbH, Walldorf, BW, GER), RapiGest™ surfactant (Waters Corporation, Milford, MA, USA), PPS Silent Surfactant (Agilent Technologies, Santa Clara, CA, USA) as well as Progenta (Protea Biosciences, Morgantown, MV, USA), which degrade proteins in combination with heat or in the low pH range [50]. For example, Abe et al. diluted a Nivolumab-containing sample with RapiGest™ surfactant and denatured it by heating it above 80 °C [36]. Also, organic solvents were used at specific concentrations for enrichment by precipitation as well as sample denaturation [30] without a noticeable reduction in enzyme activity [46]. Each of the mentioned methods has its drawbacks and needs to be optimized experimentally.
The reduction step during denaturation is essential for allowing the proteolytic enzyme to access the entire protein structure. Dithiothreitol (DTT) is commonly used as a reduction agent [30,36,38,45,51]. As an alternative, tris(2-carboxyethyl)phosphine (TCEP) was used [52,53]. TCEP had higher pH as well as temperature stability and faster-reducing properties as compared to DTT. In the context of bottom-up sequencing, it was recently shown that the use of different reducing agents had only a minor impact on peptide identification [54]. By contrast, the influence of different alkylation agents on peptide yield was apparently larger. Alkylation agents derivatize free thiol groups of cysteines, thereby inhibiting their ability to reform disulfide bonds. The most frequently used alkylation agent for antibody quantification is iodoacetamide (IAM) [36,38,51,53]. The reaction is carried out in the absence of light and at room temperature to avoid side reactions. Over the past few years, different alkylation reactions have been investigated in detail. Mueller and Winter compared IAM as an alkylation agent against iodoacetic acid (IAA), acrylamide (AA) and chloroacetamide (CAM) [55]. The authors identified unspecific reactions on the side chains of tyrosine, serine and threonine of digested HeLa proteins when using the iodine-containing reagents. Contrary to this, another study reported a higher yield of alkylated cysteine peptides and fewer side reactions when the procedure was performed using IAA compared to AA, N-ethylmaleimide or 4-vinylpyridine [54]. However, investigations by Kuznetsova et al. revealed that carbamidomethylation may affect up to 80% of peptides containing methionine after IAM-alkylation of digested proteins from HeLa and HepG2 cells [56]. Robinson and Hains have found that less light-sensitive chloroacetamide has an adverse impact on methionine oxidation [57]. Furthermore, IAM increased the rate of methionine-to-isothreonine conversion; structurally related IAA induced the same side reactions. Additionally, it has been demonstrated that IAM-alkylated peptides with N-terminal cysteines are prone to cyclization, which can result in a mass shift of −17 Da [58]. Consequently, optimization of reduction and alkylation conditions appears fundamental for antibody quantification. Due to the time-intensive optimization procedure as well as possible by-products, there are many protocols that skip these steps [28,29,30,35,37]. However, this can lead to incomplete protein digestion, as the antibody only partially unfolds or even refolds, and disulfide-connected peptides complicate the software-supported evaluation [59].

2.1.3. Digestion

Efficient enzymatic breakdown of therapeutic mAbs into peptides is crucial for reliable determination of the antibody amount by this approach. Specifically, digestion efficiencies appear to depend on the individual protein structure, in particular regarding phosphorylation and glycosylation patterns [59,60,61]. Proteases should be of the highest “MS grade” quality to ensure high specificity, activity, and purity. For MS analysis, peptides with basic residues at the C-terminus are preferred due to their enhanced ionization efficiency in positive electrospray ionization (ESI+) mode. Therefore, it is not surprising that the serine protease trypsin is the most frequently used proteolytic enzyme. Several trypsin products from different suppliers are available on the market (Table 2), varying in activity and optimal digestion conditions. It should be noted that naturally obtained trypsin from porcine or bovine pancreas can contain impurities of chymotrypsin. To prevent the protease activity of chymotrypsin, the tosyl-phenylalanyl-chloromethyl-ketone (TPCK) is added to most commercial trypsin products. Furthermore, autolysis of trypsin results in the formation of pseudotrypsin with a chymotrypsin-like activity. Both drawbacks can be avoided by using recombinant and modified trypsin, whose dimethylated lysine residues prevent self-digestion and increase stability [62,63]. Surface-immobilized trypsin is increasingly being implemented for antibody digestion because it offers faster digestion with less self-digestion [36,64] and can be integrated as immobilized enzyme reactors for automated sample processing [65]. Nevertheless, non-oriented immobilization may also result in a reduction in the accessibility of the enzyme’s active site [66] and higher quantification limits in comparison to in-solution digestions [65].
Trypsin cleaves proteins specifically at the C-terminus of the basic amino acids arginine (R) and lysine (K) (Figure 3), typically generating peptides of a size favorable for downstream HPLC–MS/MS analysis. Both amino acids are abundant and well-distributed in different proteins, producing peptides with an average length of 14 amino acids in length and a minimum of two positive charges [59]. However, when a proline residue follows at the C-terminus of R or K, the protein backbone is almost unaffected by trypsin. Furthermore, the presence of glutamic and aspartic acid, as well as phosphorylated threonine and serine, may increase the frequency of so-called missed cleavages. Therefore, some providers offer protease products combining standard trypsin with the digestion of Lys-C (Table 2), which shares C-terminal K as the cleavage site but continues to work under high urea concentrations (6–8 M urea). The combination minimizes missed K cleavages, resulting in increased digestion efficiency and improved reproducibility [67].
Sometimes, trypsin fails to generate peptides of optimal length due to the high abundance of R and K in the antibody sequence. In such cases, the use of other proteases like chymotrypsin, AspN, GluC, and ArgC can be good alternatives [68,69]. Here, the nomenclature of these enzymes corresponds to the target amino acid and terminus at which the protein is cleaved (Figure 3). Generally, enzymatic digestion should be performed in a buffer with a compatible pH that is optimal for the specific enzyme. For example, the listed products in Table 2 recommend the use of ammonium bicarbonate or Tris buffers with a pH between 7 and 8. However, Tris enhances the formation of various adduct ions and can cause ion suppression during ESI [49]. The selection of an appropriate buffer can also influence the generated peptides, whereby spontaneous chemical deamination of asparagine and glutamine residues may occur [70,71].
Furthermore, enzymatic proteolysis can be performed under different temperatures, digestion times and enzyme-to-antibody ratios. Typically, digestion with trypsin is carried out at 37 °C for one to 24 h (Table 2). Some optimized or immobilized trypsin variants are stable at higher temperatures and enable faster digestion [59]. Additionally, the incubation time can depend on the position of target signature peptides and their steric accessibility. Peptides, especially from CDRs located at the surface, are easily accessible and allow fast enzymatic digestion [72]. Lastly, prolonged digestion time and elevated temperatures can introduce unwanted protein modifications and amino acid changes like asparagine deamidation and N-terminal glutamine cyclization [70,71,73], as well as oxidation [74]. For enzymatic digestion, an enzyme-to-antibody ratio of 1:100 to 1:20 is commonly recommended. Nevertheless, it is mandatory to check digestion specificity and completeness for absolute quantification.

2.1.4. Signature Peptide Selection

Quantification of mAbs on a peptide level requires the selection of suitable signature peptides that allow both selective and sensitive detection of the target antibody in the sample matrix (Table 1). The signature peptide should consist of a unique amino acid sequence for the desired antibody and have good chromatographic separability and ionization efficiency for HPLC-MS/MS analysis. Knowledge of the amino acid sequence of the target antibody is advantageous for this selection. Generally, protein sequences of FDA-approved mAbs are available in the IMGT® (ImmunoGenetics Information system, http://www.imgt.org/ (accessed on 7 August 2024)) or databases of ABCD (AntiBodies Chemically Defined, https://web.expasy.org/abcd/ (accessed on 7 August 2024)). For the prediction of potentially generated peptides, the free programs MS-DIGEST from Protein Prospector (http://prospector.ucsf.edu (accessed on 7 August 2024)) or the deep learning program DeepDigest [75] can be used. When quantifying antibodies in human serum, peptides of the CDR are preferable due to their uniqueness for each mAb as well as enzymatic accessibility. Accordingly, predicted peptide sequences can be analyzed with the Basic Local Alignment Search Tool (BLAST, https://blast.ncbi.nlm.nih.gov/Blast.cgi (accessed on 7 August 2024)) for proteins or Peptide Atlas (http://www.peptideatlas.org (accessed on 7 August 2024)) for matches with interfering peptide sequences of the human proteome. For the quantification of purified therapeutic antibodies, choosing peptides from the constant Fc-region can also be used, allowing the quantification of multiple mAbs of similar subclass with the same MS assay.
Nevertheless, the selection of suitable signature peptides for quantitative measurements can be affected by some analytical and equipment limitations. Initially, the selected peptides should have a length of 8 to 25 amino acids. Often, shorter sequences are not unique, while longer sequences usually lead to a significant charge distribution [69]. In addition, the amino acids contained in the peptide candidates should be considered based on their susceptibility to chemical modifications and instability that have become known in recent years [76,77]. For example, cysteine, methionine, tryptophan, and histidine are well-known amino acids prone to oxidation and leading to a mass shift of +15.9949 Da. Furthermore, asparagine or glutamine can undergo deamidation during trypsin digestion, causing mass shifts of −17.0265 Da and +0.9840 Da, respectively. The loss of ammonia through N-terminal glutamine cyclization induced by prolonged digestion time also leads to a loss of −17.0265 Da. It should also be noted that therapeutic antibodies can undergo oxidation or deamination during production, purification or storage and may, therefore, already be present before enzymatic digestion [76]. Especially for pharmacokinetic quantification studies of antibodies, biotransformation effects like blood oxidation, deamidation or proteolytic degradation should also be considered [39]. In this context, signature peptides that are rich in potentially modifiable amino acids should be avoided. Other important tips for selecting the right signal peptides for targeted protein quantification have been discussed elsewhere [78,79,80].
Likewise, peptides potentially carrying glycan or phosphorylation residues should be treated with care. Falck et al. have shown that the conserved glycopeptide EEQYNSTYR of IgG1 antibody contained a high mannose content or was present as a sialylated or more complex glycoform, and a statistically significant trypsin cleavage rate was observed. This peptide should, therefore, be excluded from analysis due to unreliable quantification. [60]. Software tools such as NetNglyc [81] (https://services.healthtech.dtu.dk/services/NetNGlyc-1.0/ (accessed on 7 August 2024)), NGlyc [82] (https://github.com/bioinformaticsML/Ngly (accessed on 7 August 2024)), or the newer deep neural network-based approach DeepNGlyPred [83] (https://github.com/dukkakc/DeepNGlyPred (accessed on 7 August 2024)) can be used to check peptide sequences for potential glycosylation issues. Moreover, Solari et al. mentioned ionization and digestion issues with phosphopeptides for protein quantification in their study and recommended utilizing higher trypsin-substrate ratios or dephosphorylation prior to HPLC-MS/MS analysis to address these challenges [61].

2.2. Quantification of Intact Antibodies

In contrast to peptide-based quantification, this strategy enables the direct measurement of intact antibodies without digestion and minimal sample preparation. Due to the high molecular weight of antibodies, the quantification occurs using high-resolution mass spectrometry (HRMS) or quadrupole mass analyzers. For quantification, the antibody sample is spiked with an intact SIL analog standard (Section 3.1) in a known amount. By comparing the peak signals of the target antibody to the SIL standard, the exact concentration of the intact antibody in the sample can be determined.
This approach has proven particularly useful in pharmacokinetic studies to monitor the biotransformation of therapeutic mAbs in patients. Jian et al. published a method for absolute quantification of intact mAbs in plasma, using an isotope-labeled mAb as an internal standard and automated software-assisted mass peak deconvolution [84]. Selective preconcentration and sample clean-up of targeted antibodies from the interfering matrix were achieved by immunoaffinity capture (IAC). Similarly, a ligand binding assay (LBA) was integrated into a high-resolution (HR)-MS workflow for simultaneous quantification of different human IgG1s at intact level, including an isotopic-labeled variant as internal standard [85]. The authors validated the method regarding selectivity, sensitivity, accuracy and precision, carryover, dilution linearity as well as reproducibility and found similar performance compared to peptide-based quantification. In addition, deglycosylation of intact human IgG1 with the enzyme PNGase F resulted in a less complex full-scan MS spectrum and increased the signal for each charge state. Notably, the use of PNGase F also allows the parallel quantification of N-glycan structures, which are involved in many biological processes. Methodological concepts for this purpose have been discussed elsewhere [86,87].
However, absolute quantification of intact mAbs by HPLC-MS/MS involves a range of drawbacks. The main problem relates to the high molecular weight of antibodies [88]. Additionally, the molecular heterogeneity due to post-translational modifications (PTMs) and chemical modifications, especially O-/N-glycosylation but also phosphorylation, deamidation and oxidation, increases the complexity of the analysis [89]. Compared to peptide-level analysis, the combination and varying location of such modifications results in more and partially interfering analyte signals that need to be monitored. As the complexity of charge state and isotope distributions increases with mass in ESI mass spectra, signal intensity decreases, resulting in a loss of analytical sensitivity. Although deconvoluted spectra can circumvent this problem to some degree [90,91], their use for quantification purposes is currently not recommended due to potential issues in data processing [88]. Furthermore, the necessary enrichment of mAbs with IAC prior to HPLC-MS/MS analysis adds additional complexity and has been critically reviewed by Zhao et al. [92] If IAC enrichment is unavoidable and the SIL internal standard is used for quantification, its addition to the matrix prior to sample preparation is compulsory [93]. For quantification of intact mAbs, the commercially available isotope labeled human IgG SILu™ mAb has been applied in a few studies [84,94]. Furthermore, the mAbXmise kit from Promise Proteomics (Table 2) also contains SIL-mAbs for frequently occurring antibodies in the field of inflammatory and oncological therapies, e.g., Adalimumab [32,95]. Similarly, in-house production of SIL analogs of targeted mAbs was also investigated [85].

2.3. Quantification of Hydrolysed Antibodies

Amino acid analysis (AAA) refers to a broadly used, absolute quantification method for targeting amino acids hydrolytically released from proteins and peptide samples. Commonly, hydrolysis of 1 to 10 nmol of protein is carried out under acidic as well as oxygen-free conditions using 6 M hydrochloric acid (HCl), elevated temperatures between 90 to 150 °C and incubation times ranging from 1 to 72 h [96,97,98]. To reduce reaction time, the application of microwave-assisted heating is widely used [98]. However, substances like salts, metal ions, nutrients or hydroxyl radicals in the hydrolysis solution can potentially affect the release of certain amino acids [96,99,100]. For example, iron, copper, but also fat can lower tyrosine recoveries but can be removed by an additional SPE cleaning step. Phenol, by contrast, can prevent amino acid degradation and halogenation of tyrosine during the hydrolysis process [96,98].
The preferred HPLC modes used for AAA of hydrolyzed proteins are hydrophilic interaction chromatography (HILIC), ion exchange (IEX) or reversed phase (RP) chromatography [99]. Conventionally, AAA included a derivatization step to optimize chromatographic separation as well as UV sensitivity for UV/VIS detection. With advances in HPLC-MS/MS analysis, accurate quantification of underivatized amino acids has now become widespread. The omission of derivatization is beneficial, as it eliminates the risk of potential contamination that may arise during the derivatization process [101].
Analogous to the absolute quantification of peptides, HPLC-MS/MS allows absolute AAA by including SIL internal standards in the analysis. Here, SIL internal standards can be full-length protein analogs [102] (Section 3.1), peptides (Section 3.2) or amino acids [103,104] (Section 3.3). By comparing the peak signals of the natural amino acids to those of the SIL standards, the exact concentration of selected amino acids from the antibody can be determined. When certified reference materials (CRMs) are used, measurements become fully traceable to the international standard system of units (SI units) [98]. This system ensures consistency and standardization in measurements around the world, making it easier for people to share and compare data. By using four SIL amino acids as internal standards in an HPLC-MS/MS setup, Jeong et al. developed an accurate quantification method for human growth hormone with an intra- and inter-day precision of relative standard deviation (RSD) of less than 1% [105]. Accordingly, the absolute quantification of proteins by AAA allows traceable and comparable results, as well as the certification of therapeutic protein products with certified SIL reference materials. Interlaboratory comparisons for therapeutic antibody quantification [106] have proven the validity of this technique. Furthermore, this approach has also contributed to the development of several protein reference materials, such as COVID-19 analytical reagents [103,104,107].

3. Selection of Internal Standards for Quantification with HPLC-MS/MS

The choice of suitable internal standards is critical for quantitative MS, as losses during digestion and hydrolysis, as well as matrix effects, can lead to significant technical bias in quantification. SIL internal standards contain at least one stable isotope (usually 13C, 15N or D) in at least one amino acid, which usually ensures co-elution with the respective target peptide or amino acid. Due to the known concentration of the added SIL internal standard, the unknown concentration of antibodies in the sample can be determined based on the signals in the HPLC-MS/MS analysis (Figure 4). Accurate quantification of mAbs was demonstrated using both SIL proteins and peptides, which in turn can be quantified by means of SIL peptides or amino acid standards. To enable metrological comparability and reliability of measurement results, calibration should be performed using SIL internal standards that can be traced back to reference materials.

3.1. Intact Antibody Standards

Different SIL antibody standards were presented for mAb quantification at the intact level. Mainly, commercially available SIL-mAbs were used and offered from different suppliers like Promise Proteomics [108] or Sigma Aldrich. The use of full-length [32,37] or partial [32,37] SIL analogs for digested mAbs were also tested and included in quantification protocols. These internal standards can be calibrated using reference materials of known quantity, the so-called protein standard absolute quantification (PSAQ) method [102,109]. For example, Lebert et al. demonstrated simultaneous quantification of several therapeutically relevant and structurally similar IgG1 and IgG4 isotypes using PSAQ [52]. For in-house production of SIL mAbs, the antibody-producing cell line is allowed to grow in media containing isotopologues of one or more amino acids, which are incorporated into the desired mAb [110]. Although SIL amino acids are readily available, the approach is usually cost- and time-intensive [93].
A more cost-effective alternative for peptide-based quantification with antibody standards is to use SIL mAbs that produce identical or similar peptides in enzymatic digestion, allowing multiple therapeutic antibodies to be quantified simultaneously. For instance, the quantification of the therapeutic antibodies infliximab, alemtuzumab, and bevacizumab using SIL mAb analogs showed sufficient precision, i.e., coefficient of variation (CV) of less than 20%, when signature peptides had a similar amino acid sequence and were located in the VH- and VL-region [111]. Furthermore, the purchasable human IgG1 SILuMAb with SIL arginine and lysine residues can also be a less expensive alternative for peptide-based quantification due to a common FC region with other therapeutic mAbs. However, as noted by Smit and co-workers, the digestion kinetics can vary between SIL mAbs and the mAbs of interest when different recombinant expression systems were used, likely due to associated variations in post-translational modifications [112].
In summary, internal standardization with SIL mAbs can provide crucial advantages in terms of reproducibility and robustness. Due to the identical behavior of targeted and SIL antibodies, throughout the entire analytical workflow, variability and inferences in MS analysis can be reduced to a minimum. However, a significant drawback is the overall cost and quality of SIL antibody standards, which currently restricts their broader application.

3.2. Peptide Standards

SIL peptide standards generally share the same AA sequence with the selected signature peptides. SIL peptides can be easily synthesized in HPLC-grade purity and are thus much cheaper than intact SIL mAbs. SIL peptide-based mAb quantification has become common practice over the last decade [36,42,51,53]. An important factor is the concentration and purity of SIL peptides that should be verified by AAA to obtain precise reference values [98,113]. Burkitt et al. established a method for protein quantification in which the SIL peptide standards were quantified by isotope dilution MS (IDMS) [114]. By using certified amino acid reference materials, mAb quantification was fully traceable to SI units.
In contrast to antibody standards, SIL peptides can only be corrected for variability in instrumental factors and matrix effects but not for changes in sample purification or enrichment prior to digestion. Extended SIL peptides contain additional amino acids adjacent to the cleavage site of the signature peptide. They can improve tracing digestion efficiency as additional amino acids need to be eliminated during enzymatic cleavage. Benesova et al. demonstrated that SIL peptides extended by three amino acids at each N- and C-terminus enable quantification results for albumin equivalent to those based on a SIL protein [115]. However, digestion kinetics can also differ for targeted mAbs and extended SIL peptides due to the accessibility of the cleavage site. For example, Li et al. obtained higher precision with a SIL intact antibody (<16% CV) than with SIL peptides in both standard and extended variants (>25%) [116]. Therefore, it is often recommended to measure at least two signature peptides to compare cleavage rates and to produce more robust results [42,117].
Peptide modifications occurring during sample preparation are another factor that can prevent accurate quantification of mAbs. For instance, CDR-specific SIL peptide standards prone to methionine oxidation impaired the quantification of therapeutic mAbs [74]. Similarly, the quantification of growth hormone indicated that the addition of the SIL peptide prior to digestion was crucial for correcting peptide degradation rates [117].
Because IgG1 and IgG4 are the primary subclasses used for therapeutic antibodies (Table 1), SIL peptides of either the constant heavy or constant light chain have the advantage of being applicable for quantification of multiple therapeutic mAbs [118,119].

3.3. Amino Acid Standards

As previously described, only a subset of the 20 proteinogenic amino acids have proven to be sterically accessible and generally stable enough for the acidic hydrolysis procedure. For example, peptide bonds between aliphatic/hydrophobic amino acids are difficult to break, resulting in insufficient release and underestimation without additional hydrolysis time. Furthermore, the chemical oxidation of methionine or deamination of asparagine and glutamine into their acid counterparts makes these amino acids unsuitable for quantification. Although correction factors were developed, the extent of hydrolysis losses and degradation artifacts were shown to vary between laboratories. Therefore, absolute protein quantification by AAA is currently limited to specific amino acids, with proline, valine, isoleucine, leucine, and phenylalanine being favored in the literature [103,104,106,107,114].

4. Software Tools Supporting Targeted mAb Quantification

The commonly used acquisition mode for absolute mAb quantification at the peptide or amino acid level is multiple reaction monitoring (MRM), using highly sensitive and selective QqQ or quadrupole-ion trap (Q-Trap) mass spectrometers. Although targeted peptides or amino acids and their spiked SIL analogs exhibit nearly identical physicochemical properties (e.g., retention time and fragmentation patterns), the MS1 monoisotopic mass shifts depending on the incorporation of labeled isotopes. Quantification based on MRM involves extracted ion chromatograms (XICs) that are created for both the precursor and product m/z of the selected peptides or amino acids. By relating peak areas of added SIL standard to those of the target peptides or amino acids, the antibody concentration can be precisely determined. The development of efficient MRM schemes depends on many critical factors such as chromatographic separation, selectivity of transitions, as well as fine-tuned fragmentation energies. Furthermore, metrological parameters, including a limit of detection and quantification (LOD, LOQ), accuracy, precision, linearity, and stability, following FDA [120] or European Medicines Agency (EMA) [121] guidelines, must be taken into consideration during method development. Necessary procedures for the latter have already been discussed in detail for beginners elsewhere [79,122]. Here, we focus on commercial and open-source software packages that support method development and data analysis but can themselves pose a challenge for inexperienced users in the field (Table 3). Software tools enabling time-efficient and user-friendly processing of large sample numbers obtained from different HPLC-MS/MS setups are also discussed. Furthermore, solutions for the automated prediction of tryptic peptides and their organization into spectral libraries are addressed.

4.1. Commercial and Device-Specific Software

All leading MS manufacturers offer workflow-oriented software solutions supporting mAb characterization, partially in combination with pre-configured instrument stacks, such as the BioAccord System (Waters Corporation, Milford, MA, USA). The latter is specifically designed for routine and automated monitoring of biotherapeutics in quality control processes, including multi-attribute method (MAM) workflows for analyzing antibody degradation and modification, e.g., during storage [123] and quantification of mAbs at the intact level [124]. More general software for MRM analysis by the same manufacturer includes the TargetLynx™ Application Manager (method development, automated sample data acquisition, processing, and reporting) and QuanOptimize™ (automated optimization of MRM transitions and collision energies). Similarly, BioPharma FinderTM (Thermo Fisher Scientific, Waltham, MA, USA) is a comprehensive software package offering pre-defined workflows, e.g., enabling quantification at the intact level due to meticulous deconvolution of mass spectra or multi-attribute characterization of mAbs in contexts such as bioprocessing up to final product quality control [35,125,126]. To identify proteotypic peptides and predict fragmentation patterns for targeted quantification, PinpointTM (Thermo Fisher Scientific) helps to determine MRM transitions and can export methods directly to compatible instruments. Due to an iterative workflow design, acquired data can be used to refine acquisition methods and verify top peptide candidates [127,128]. MassHunter (Agilent Technologies, Santa Clara, CA, USA) includes various packages for data acquisition and quantitative analysis. MassHunter Optimizer supports the development of MRM methods by automatically optimizing acquisition parameters, facilitating, e.g., the development of more sensitive triggered MRM (tMRM) methods [129]. The Analyst® software suite (version 1.7.3, AB Sciex, Framingham, MA, USA) not only provides general MS data acquisition and analysis but also offers a variety of tools specifically designed for the analysis of mAbs. These tools include intact mass spectrum characterization, subunit analysis, peptide mapping, glycan analysis, and drug-antibody ratio (DAR) calculations (Biologics Explorer, ProteinPilotTM). In addition, MRMPilotTM supports building and optimizing peptide MRM experiments, allowing inspection of full scan MS/MS data as well as transferring transition settings to the acquisition module. MRMPilotTM also allows input from open databases such as PeptideAtlas [130]. Similarly to MassHunter, it also supports the developing of MRM-triggered acquisition methods. For data evaluation, MultiQuantTM (AB Sciex) supports absolute quantification based on multiple peptides and the creation of reports including common quality parameters such as the CV and accuracy.
Furthermore, third-party software primarily aimed at high-throughput proteomics can be applied to targeted quantification tasks. Available options include SpectronautTM, SpectromineTM, and SpectroDiveTM (Biognosys, Schlieren, DIE, ZH), as well as Mascot Distiller (Matrix Science, Mount Prospect, IL, USA). These software packages accept data from all major MS instruments.

4.2. Open-Source Software Alternatives

In addition to commercially available software solutions, several open-source programs and web-based applications have become available for MRM method development and data analysis. One of the most popular options covering the entire experimental workflow is Skyline, which was released by the MacCoss group in 2009 [131]. The software is manufacturer-independent and is designed for the quantification of small molecules, including amino acids and peptides. The software enables the comparison of MS/MS spectra with integrated databases and the development of targeted MRM methods. The functionality of the software is subject to continuous improvement based on user feedback. It includes the prediction of peptide ions, and their transitions based on the uploaded antibody sequence, selected modifications (isotopic labels, PTMs), digestion enzyme and instrument-specific requirements. Furthermore, the software enables the prediction of retention times and facilitates the construction of spectral libraries with the machine learning program Prosit [132]. A range of free tutorials provide entry points for new users to the various aspects of data analysis.
Another freely available and widely used alternative for data evaluation is MaxQuant, developed by groups around Jürgen Cox and Matthias Mann at the Max Planck Institute of Biochemistry [133]. Analogous to Skyline, it is designed to analyze high-throughput mass-spectrometry data in a desktop application. It supports various labeling techniques and algorithms for peak detection, as well as peptide identification with corresponding MS2 identification (Andromeda). However, it is more suitable for HRMS data than for MRM data. A substantial collection of published protocols is available for the implementation of MaxQuant in one’s own pipeline for the analysis of mass spectrometric data [134,135].
In addition to the previously discussed options, there are other, more sophisticated software solutions that will be briefly outlined here. OpenMS differs from Skyline in that it provides a flexible programming framework (Python) rather than a ready-to-use desktop interface [136,137]. Functionality includes core algorithms for MS data analysis, such as peak detection, alignment, integration, etc., as well as predefined high-level workflows. MRMPROBS, again, is a stand-alone desktop application aimed at the automatic detection and identification of MRM signals based on probabilistic criteria [138]. Features include XIC panels for efficient quality control (QC) and manual peak correction, but no direct support for peptide targets is provided; target lists must be provided as a text file. MassChroQ is a command-line program supporting both low-resolution (MRM) and HRMS data, as peak quantification is based on XICs rather than feature detection [139]. Peaks from all samples are automatically aligned and exported to a spreadsheet format, allowing integration into data pipelines. MRMAnalyzer similarly focuses on automated pipelines but is implemented as an R package [140,141]. Additionally, MassIVE.quant [142], quantms [143], and MRMPro [144] are examples of recent cloud-based platforms for quantitative proteomics, featuring large-scale online data processing capabilities and extensive QC plots for peak inspection. In the future, artificial intelligence and deep learning algorithms will likely support the user comprehensively by accurately predicting fragmentation patterns and providing “smart” data analysis tools for proteomics in general [145]

5. Outlook: Need for Standardized Protocols, Certified Reference Materials, and New Technologies

The use of recombinant antibodies in both therapeutic and diagnostic applications will continue to increase over the next few years. Their exact quantification, both in pure form and in serum, plasma, or blood samples, is therefore essential. As shown in the previous chapters, a variety of peptide-based methods and protocols have been developed in recent decades with different digestion conditions or SIL materials and for each commercial antibody individually (Table 1). It is, therefore, not surprising that different methods can lead to different results, making it difficult for inexperienced users to select a suitable method for the quantification of antibodies. In the authors’ view, standardization of the methods would facilitate more comparable results across research institutions, industry, and clinical studies. In contrast, achieving harmonization regarding the HPLC-MS/MS measurement technology and analysis software used is considered to present a significant challenge.
The need for standardized methods was impressively demonstrated in a study about the quantification of a recombinant human IgG1 SARS-CoV-2 antibody by the Protein Analysis Working Group (PAWG) of the Comité Consultatif pour la Quantité de Matière (CCQM) [106,146]. Participating national metrology institutes (NMIs) showed that there was a large discrepancy in the results (up to 22% difference from reference value) despite the use of isotope dilution mass spectrometry (ID-MS) for peptide-based quantification approaches. Based on this, the digestion conditions and peptide selection for this SARS-CoV-2 antibody were optimized and validated with a reference antibody and AAA as reference method [53]. The authors clearly demonstrated how important it is to use CRMs to develop robust methods and generate accurate results. The need for suitable reference materials and standardized measurements is also emphasized in the clinical field for the quantification of biomarkers, e.g., apolipoprotein A [147,148,149]. Precise quantification is crucial in this context to enable accurate monitoring of biomarkers and effective patient treatment. Furthermore, the use of CRMs makes the results more comparable between different clinical facilities. The implementation of reference materials for quantitative HPLC-MS/MS assay development enables metrological traceability of calibration standards, ensuring that measurement results have a documented and unbroken chain of traceability to SI units [112]. Furthermore, it can also compensate for differences in the device-specific ionizations or software-dependent peak detection and smoothing settings.
A broadly accessible metrological reference material would provide a representative standard to enhance the harmonization of antibody quantification measurements [150]. For the development of standardized methods, the two certified antibody reference materials NISTmAb (RM 8671) and AISTmAb (RM 6208-a) from the National Institute of Standards and Technology (NIST, USA) and National Institute of Advanced Industrial Science and Technology (AIST, Tokyo, Japan) are available [151,152]. Due to the extensive effort needed to establish “certified” values, only a limited number of analytes are certified as reference material. The necessity for CRMs has also become especially evident during the COVID-19 pandemic, particularly in the context of validating analytical tests [103,104,107]. Additionally, the World Health Organization (WHO) also established several antibody reference standards, including the WHO Adalimumab reference standard (ref. 17/236) and the WHO infliximab reference standard (ref. 16/170). However, these standards exhibit inaccuracies in their concentrations [32], highlighting the need for certified, traceable materials that should be characterized through multidisciplinary approaches and evaluated by various laboratories [153]. When selecting suitable calibrators, it is essential to consider the differences in PTMs, as these variations can lead to potential deviations in measurement results [154].
Apart from method standardization and CRM development, new sample preparation technologies may also contribute to faster and more streamlined quantification assay development. We regard innovations in two fields as particularly promising. The first field relates to innovative enzyme technologies for the optimization of protein digestion. A recent method that employs aqueous microdroplet-mediated enzymatic digestion has shown a remarkable decrease in digestion times, achieving results in the millisecond range, in contrast to the several hours typically required by conventional methods [155]. Moreover, this approach has achieved a very high sequence coverage. Similarly, recombinant and immobilized enzyme systems have demonstrated potential in boosting cleavage rates, apparently due to improved stability, inhibition of trypsin self-digestion and higher enzyme-to-protein ratios [59,63]. Furthermore, there are also initial publications on thermostable enzyme variants, for example by inserting mutations or glycosylation [156,157]. We believe that significant further optimization of enzyme systems is feasible due to progress in biotechnology, material science and artificial intelligence (AI)-supported designs. Thereby, the dependency of digestion efficiency on the primary sequence and PTMs of mAb targets could be minimized, resulting in much-improved commutability of the calibrator and SIL materials.
A second field where innovation can have a significant impact on future mAb assays pertains to the production of SIL mAbs. The recently announced mAbXmise kit [95] offers a convenient mixture of several SIL mAbs, enabling the simultaneous quantification of various therapeutic mAbs [32]. The development of further mAb kits may result in more efficient production pipelines, thereby enhancing the broader and more affordable availability of SIL mAbs. This approach could improve compensation for parameters such as digestion conditions and purification from complex matrices, leading to more reproducible results. Ideally, an increase in demand could lead to the commercial availability of SIL mAbs becoming comparable to that of SIL peptides. However, production is expected to remain more complex and costly in the foreseeable future. Cell-free in vitro systems, such as PURE (Protein synthesis Using Recombinant Elements), could provide a cost-effective solution for the production of SIL mAbs [158].

Author Contributions

Conceptualization and review writing, S.D.; conceptualization, writing, and supervision, Z.K. and C.J.; critical reading and editing, M.G.W. and Y.R. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

Special thanks to Gaby Bosc-Bierne for critical reading and remarks.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
AAAcrylamide
AAAAmino acid analysis
ABCDAntiBodies Chemically Defined
ADCAntibody-drug conjugate
AIArtificial intelligence
AISTNational Institute of Advanced Industrial Science and Technology
BLASTBasic Local Alignment Search Tool
CAMChloroacetamide
CDRComplementarity-determining region
CH1-3Constant heavy chains 1 to 3
CHOChinese hamster ovary
CL Constant light chain
CRMCertified reference material
CVCoefficient of variation
DARDrug-antibody ratio
DOCSodium dodecyl sulfate
DTTDithiothreitol
EMAEuropean Medicines Agency
ESIElectrospray ionization
FABAntigen-binding sites of antibody
FCFragment crystallizable region
FDAFood and Drug Administration
HAMAHuman anti-mouse antibody
HILICHydrophilic interaction chromatography
HPLC-MS/MSHigh-performance liquid chromatography-tandem mass spectrometry
HRMSHigh-resolution mass spectrometry
IAAIodoacetic acid
IACImmunoaffinity capture
IAMIodoacetamide
ID-MS Isotope dilution mass spectrometry
IEXIon exchange
IgG1Immunoglobulin G antibody class 1
IgG4Immunoglobulin G antibody class 4
IL-12Interleukin-12
IL-23Interleukin-23
IMGTImmunoGenetics Information system
KLysine
LBALigand binding assay
LODLimit of detection
LOQLimit of quantification
mAbTherapeutic monoclonal antibody
MAMMulti-attribute method
MRMMultiple reaction monitoring
MSMass spectrometry
NISTNational Institute of Standards and Technology
NMINational Metrology Institute
opt.Optional
PD-1Programmed cell death protein 1
PSAQProtein standard absolute quantification
PTMPost-translational modification
PUREProtein synthesis Using Recombinant Elements
QCQuality control
QqQTriple-quadrupole
Q-TrapQuadrupole-ion trap
RArginine
RPReversed phase
RSDRelative standard deviation
SDSSodium dodecyl sulfate
SI unitsInternational standard system of units
SILStable isotope-labeled
SPESolid-phase extraction
TCEPTris(2-carboxyethyl)phosphine
tMRMTriggered MRM
TNF-αTumor necrosis factor-alpha
TPCKTosyl-phenylalanyl-chloromethyl-ketone
VHVariable heavy chain
VLVariable light chain
WHOWorld Health Organization
XICExtracted ion chromatogram

References

  1. Köhler, G.; Milstein, C. Continuous cultures of fused cells secreting antibody of predefined specificity. Nature 1975, 256, 495–497. [Google Scholar] [CrossRef] [PubMed]
  2. Todd, P.A.; Brogden, R.N. Muromonab CD3. A review of its pharmacology and therapeutic potential. Drugs 1989, 37, 871–899. [Google Scholar] [CrossRef] [PubMed]
  3. Schroff, R.W.; Foon, K.A.; Beatty, S.M.; Oldham, R.K.; Morgan, A.C., Jr. Human anti-murine immunoglobulin responses in patients receiving monoclonal antibody therapy. Cancer Res. 1985, 45, 879–885. [Google Scholar] [PubMed]
  4. Jaffers, G.J.; Fuller, T.; Cosimi, A.; Russell, P.; Winn, H.; Colvin, R. Monoclonal antibody therapy. Anti-idiotypic and non-anti-idiotypic antibodies to OKT3 arising despite intense immunosuppression. Transplantation 1986, 41, 572–578. [Google Scholar] [CrossRef]
  5. Morrison, S.L.; Johnson, M.J.; Herzenberg, L.A.; Oi, V.T. Chimeric human antibody molecules: Mouse antigen-binding domains with human constant region domains. Proc. Natl. Acad. Sci. USA 1984, 81, 6851–6855. [Google Scholar] [CrossRef]
  6. Brüggemann, M.; Winter, G.; Waldmann, H.; Neuberger, M.S. The immunogenicity of chimeric antibodies. J. Exp. Med. 1989, 170, 2153–2157. [Google Scholar] [CrossRef]
  7. Faulds, D.; Sorkin, E.M. Abciximab (c7E3 Fab). A review of its pharmacology and therapeutic potential in ischaemic heart disease. Drugs 1994, 48, 583–598. [Google Scholar] [CrossRef]
  8. Leget, G.A.; Czuczman, M.S. Use of rituximab, the new FDA-approved antibody. Curr. Opin. Oncol. 1998, 10, 548–551. [Google Scholar] [CrossRef]
  9. Jones, P.T.; Dear, P.H.; Foote, J.; Neuberger, M.S.; Winter, G. Replacing the complementarity-determining regions in a human antibody with those from a mouse. Nature 1986, 321, 522–525. [Google Scholar] [CrossRef]
  10. Queen, C.; Schneider, W.P.; Selick, H.E.; Payne, P.W.; Landolfi, N.F.; Duncan, J.F.; Avdalovic, N.M.; Levitt, M.; Junghans, R.P.; Waldmann, T.A. A humanized antibody that binds to the interleukin 2 receptor. Proc. Natl. Acad. Sci. USA 1989, 86, 10029–10033. [Google Scholar] [CrossRef]
  11. Wiseman, L.R.; Faulds, D. Daclizumab: A review of its use in the prevention of acute rejection in renal transplant recipients. Drugs 1999, 58, 1029–1042. [Google Scholar] [CrossRef] [PubMed]
  12. Safdari, Y.; Farajnia, S.; Asgharzadeh, M.; Khalili, M. Antibody humanization methods—A review and update. Biotechnol. Genet. Eng. Rev. 2013, 29, 175–186. [Google Scholar] [CrossRef] [PubMed]
  13. McCafferty, J.; Griffiths, A.D.; Winter, G.; Chiswell, D.J. Phage antibodies: Filamentous phage displaying antibody variable domains. Nature 1990, 348, 552–554. [Google Scholar] [CrossRef]
  14. Smith, G.P. Filamentous fusion phage: Novel expression vectors that display cloned antigens on the virion surface. Science 1985, 228, 1315–1317. [Google Scholar] [CrossRef]
  15. Zhang, Y. Evolution of phage display libraries for therapeutic antibody discovery. mAbs 2023, 15, 2213793. [Google Scholar] [CrossRef]
  16. Frenzel, A.; Schirrmann, T.; Hust, M. Phage display-derived human antibodies in clinical development and therapy. mAbs 2016, 8, 1177–1194. [Google Scholar] [CrossRef]
  17. Sánchez-Robles, E.M.; Girón, R.; Paniagua, N.; Rodríguez-Rivera, C.; Pascual, D.; Goicoechea, C. Monoclonal Antibodies for Chronic Pain Treatment: Present and Future. Int. J. Mol. Sci. 2021, 22, 10325. [Google Scholar] [CrossRef]
  18. Lonberg, N.; Taylor, L.D.; Harding, F.A.; Trounstine, M.; Higgins, K.M.; Schramm, S.R.; Kuo, C.-C.; Mashayekh, R.; Wymore, K.; McCabe, J.G.; et al. Antigen-specific human antibodies from mice comprising four distinct genetic modifications. Nature 1994, 368, 856–859. [Google Scholar] [CrossRef]
  19. Jakobovits, A.; Amado, R.G.; Yang, X.; Roskos, L.; Schwab, G. From XenoMouse technology to panitumumab, the first fully human antibody product from transgenic mice. Nat. Biotechnol. 2007, 25, 1134–1143. [Google Scholar] [CrossRef]
  20. Lapadula, G.; Marchesoni, A.; Armuzzi, A.; Blandizzi, C.; Caporali, R.; Chimenti, S.; Cimaz, R.; Cimino, L.; Gionchetti, P.; Girolomoni, G.; et al. Adalimumab in the Treatment of Immune-Mediated Diseases. Int. J. Immunopathol. Pharmacol. 2014, 27, 33–48. [Google Scholar] [CrossRef]
  21. Verdin, P. Top companies and drugs by sales in 2023. Nat. Rev. Drug Discov. 2024, 23, 240. [Google Scholar] [CrossRef] [PubMed]
  22. Barbee, M.S.; Ogunniyi, A.; Horvat, T.Z.; Dang, T.-O. Current Status and Future Directions of the Immune Checkpoint Inhibitors Ipilimumab, Pembrolizumab, and Nivolumab in Oncology. Ann. Pharmacother. 2015, 49, 907–937. [Google Scholar] [CrossRef] [PubMed]
  23. Liu, K.; Li, M.; Li, Y.; Li, Y.; Chen, Z.; Tang, Y.; Yang, M.; Deng, G.; Liu, H. A review of the clinical efficacy of FDA-approved antibody–drug conjugates in human cancers. Mol. Cancer 2024, 23, 62. [Google Scholar] [CrossRef] [PubMed]
  24. Panowski, S.; Bhakta, S.; Raab, H.; Polakis, P.; Junutula, J.R. Site-specific antibody drug conjugates for cancer therapy. mAbs 2014, 6, 34–45. [Google Scholar] [CrossRef]
  25. Zhu, X.; Huo, S.; Xue, C.; An, B.; Qu, J. Current LC-MS-based strategies for characterization and quantification of antibody-drug conjugates. J. Pharm. Anal. 2020, 10, 209–220. [Google Scholar] [CrossRef]
  26. Todoroki, K.; Yamada, T.; Mizuno, H.; TOYO’OKA, T. Current mass spectrometric tools for the bioanalyses of therapeutic monoclonal antibodies and antibody-drug conjugates. Anal. Sci. 2018, 34, 397–406. [Google Scholar] [CrossRef]
  27. Cahuzac, H.; Devel, L. Analytical Methods for the Detection and Quantification of ADCs in Biological Matrices. Pharmaceuticals 2020, 13, 462. [Google Scholar] [CrossRef]
  28. Millet, A.; Khoudour, N.; Guitton, J.; Lebert, D.; Goldwasser, F.; Blanchet, B.; Machon, C. Analysis of Pembrolizumab in Human Plasma by LC-MS/HRMS. Method Valid. Comp. Elisa. Biomed. 2021, 9, 621. [Google Scholar] [CrossRef]
  29. Hallin, E.I.; Serkland, T.T.; Bjånes, T.K.; Skrede, S. High-throughput, low-cost quantification of 11 therapeutic antibodies using caprylic acid precipitation and LC-MS/MS. Anal. Chim. Acta 2024, 1313, 342789. [Google Scholar] [CrossRef]
  30. de Jong, K.A.; Rosing, H.; Huitema, A.D.; Beijnen, J.H. Optimized sample pre-treatment procedure for the simultaneous UPLC-MS/MS quantification of ipilimumab, nivolumab, and pembrolizumab in human serum. J. Chromatogr. B 2022, 1196, 123215. [Google Scholar] [CrossRef]
  31. Willeman, T.; Jourdil, J.-F.; Gautier-Veyret, E.; Bonaz, B.; Stanke-Labesque, F. A multiplex liquid chromatography tandem mass spectrometry method for the quantification of seven therapeutic monoclonal antibodies: Application for adalimumab therapeutic drug monitoring in patients with Crohn’s disease. Anal. Chim. Acta 2019, 1067, 63–70. [Google Scholar] [CrossRef] [PubMed]
  32. Tron, C.; Lemaitre, F.; Bros, P.; Goulvestre, C.; Franck, B.; Mouton, N.; Bagnos, S.; Coriat, R.; Khoudour, N.; Lebert, D.; et al. Quantification of infliximab and adalimumab in human plasma by a liquid chromatography tandem mass spectrometry kit and comparison with two ELISA methods. Bioanalysis 2022, 14, 831–844. [Google Scholar] [CrossRef] [PubMed]
  33. Scheffe, N.; Schreiner, R.; Thomann, A.; Findeisen, P. Development of a mass spectrometry-based method for quantification of ustekinumab in serum specimens. Ther. Drug Monit. 2020, 42, 572–577. [Google Scholar] [CrossRef] [PubMed]
  34. Yamaoka, K.; Irie, K.; Hiramoto, N.; Hirabatake, M.; Ikesue, H.; Hashida, T.; Shimizu, T.; Ishikawa, T.; Muroi, N. Safety and blood levels of daratumumab after switching from intravenous to subcutaneous administration in patients with multiple myeloma. Investig. New Drugs 2023, 41, 761–767. [Google Scholar] [CrossRef]
  35. Li, W.; Huang, W.; Yu, X.; Chen, C.; Yuan, Y.; Liu, D.; Wang, F.; Yu, J.; Diao, X. A validated LC-MS/MS method for the quantitation of daratumumab in rat serum using rapid tryptic digestion without IgG purification and reduction. J. Pharm. Biomed. Anal. 2024, 243, 116083. [Google Scholar] [CrossRef]
  36. Abe, K.; Shibata, K.; Naito, T.; Karayama, M.; Hamada, E.; Maekawa, M.; Yamada, Y.; Suda, T.; Kawakami, J. Quantitative LC-MS/MS method for nivolumab in human serum using IgG purification and immobilized tryptic digestion. Anal. Methods 2020, 12, 54–62. [Google Scholar] [CrossRef]
  37. Millet, A.; Khoudour, N.; Bros, P.; Lebert, D.; Picard, G.; Machon, C.; Goldwasser, F.; Blanchet, B.; Guitton, J. Quantification of nivolumab in human plasma by LC-MS/HRMS and LC-MS/MS, comparison with ELISA. Talanta 2021, 224, 121889. [Google Scholar] [CrossRef]
  38. Heudi, O.; Barteau, S.; Zimmer, D.; Schmidt, J.; Bill, K.; Lehmann, N.; Bauer, C.; Kretz, O. Towards Absolute Quantification of Therapeutic Monoclonal Antibody in Serum by LC−MS/MS Using Isotope-Labeled Antibody Standard and Protein Cleavage Isotope Dilution Mass Spectrometry. Anal. Chem. 2008, 80, 4200–4207. [Google Scholar] [CrossRef]
  39. Wei, C.; Su, D.; Wang, J.; Jian, W.; Zhang, D. LC–MS challenges in characterizing and quantifying monoclonal antibodies (mAb) and antibody-drug conjugates (ADC) in biological samples. Curr. Pharmacol. Rep. 2018, 4, 45–63. [Google Scholar] [CrossRef]
  40. Hentschel, A.; Piontek, G.; Dahlmann, R.; Findeisen, P.; Sakson, R.; Carbow, P.; Renné, T.; Reinders, Y.; Sickmann, A. Highly sensitive therapeutic drug monitoring of infliximab in serum by targeted mass spectrometry in comparison to ELISA data. Clin. Proteom. 2024, 21, 16. [Google Scholar] [CrossRef]
  41. Xu, K.; Liu, L.; Maia, M.; Li, J.; Lowe, J.; Song, A.; Kaur, S. A multiplexed hybrid LC–MS/MS pharmacokinetic assay to measure two co-administered monoclonal antibodies in a clinical study. Bioanalysis 2014, 6, 1781–1794. [Google Scholar] [CrossRef] [PubMed]
  42. Chiu, H.-H.; Liao, H.-W.; Shao, Y.-Y.; Lu, Y.-S.; Lin, C.-H.; Tsai, I.-L.; Kuo, C.-H. Development of a general method for quantifying IgG-based therapeutic monoclonal antibodies in human plasma using protein G purification coupled with a two internal standard calibration strategy using LC-MS/MS. Anal. Chim. Acta 2018, 1019, 93–102. [Google Scholar] [CrossRef]
  43. Ouyang, Z.; Furlong, M.T.; Wu, S.; Sleczka, B.; Tamura, J.; Wang, H.; Suchard, S.; Suri, A.; Olah, T.; Tymiak, A. Pellet digestion: A simple and efficient sample preparation technique for LC–MS/MS quantification of large therapeutic proteins in plasma. Bioanalysis 2012, 4, 17–28. [Google Scholar] [CrossRef]
  44. Yang, Z.; Hayes, M.; Fang, X.; Daley, M.P.; Ettenberg, S.; Tse, F.L. LC− MS/MS Approach for Quantification of Therapeutic Proteins in Plasma Using a Protein Internal Standard and 2D-Solid-Phase Extraction Cleanup. Anal. Chem. 2007, 79, 9294–9301. [Google Scholar] [CrossRef]
  45. Millán-Martín, S.; Jakes, C.; Carillo, S.; Bones, J. Multi-attribute method (MAM) to assess analytical comparability of adalimumab biosimilars. J. Pharm. Biomed. Anal. 2023, 234, 115543. [Google Scholar] [CrossRef]
  46. El Amrani, M.; Gerencser, L.; Huitema, A.D.; Hack, C.E.; van Luin, M.; van der Elst, K.C. A generic sample preparation method for the multiplex analysis of seven therapeutic monoclonal antibodies in human plasma or serum with liquid chromatography-tandem mass spectrometry. J. Chromatogr. A 2021, 1655, 462489. [Google Scholar] [CrossRef]
  47. Proc, J.L.; Kuzyk, M.A.; Hardie, D.B.; Yang, J.; Smith, D.S.; Jackson, A.M.; Parker, C.E.; Borchers, C.H. A quantitative study of the effects of chaotropic agents, surfactants, and solvents on the digestion efficiency of human plasma proteins by trypsin. J. Proteome Res. 2010, 9, 5422–5437. [Google Scholar] [CrossRef]
  48. Sun, S.; Zhou, J.-Y.; Yang, W.; Zhang, H. Inhibition of protein carbamylation in urea solution using ammonium-containing buffers. Anal. Biochem. 2014, 446, 76–81. [Google Scholar] [CrossRef]
  49. Loo, R.R.O.; Dales, N.; Andrews, P.C. Surfactant effects on protein structure examined by electrospray ionization mass spectrometry. Protein Sci. 1994, 3, 1975–1983. [Google Scholar] [CrossRef]
  50. Shieh, I.F.; Lee, C.-Y.; Shiea, J. Eliminating the Interferences from TRIS Buffer and SDS in Protein Analysis by Fused-Droplet Electrospray Ionization Mass Spectrometry. J. Proteome Res. 2005, 4, 606–612. [Google Scholar] [CrossRef]
  51. Rogers, J.C.; Bomgarden, R.D. Sample preparation for mass spectrometry-based proteomics; from proteomes to peptides. In Modern Proteomics—Sample Preparation, Analysis and Practical Applications; Springer: Berlin/Heidelberg, Germany, 2016; pp. 43–62. [Google Scholar]
  52. Lebert, D.; Picard, G.; Beau-Larvor, C.; Troncy, L.; Lacheny, C.; Maynadier, B.; Low, W.; Mouz, N.; Brun, V.; Klinguer-Hamour, C. Absolute and multiplex quantification of antibodies in serum using PSAQ™ standards and LC-MS/MS. Bioanalysis 2015, 7, 1237–1251. [Google Scholar] [CrossRef] [PubMed]
  53. Martos, G.; Bedu, M.; Josephs, R.; Westwood, S.; Wielgosz, R. Quantification of SARS-CoV-2 monoclonal IgG mass fraction by isotope dilution mass spectrometry. Anal. Bioanal. Chem. 2024, 416, 2423–2437. [Google Scholar] [CrossRef] [PubMed]
  54. Suttapitugsakul, S.; Xiao, H.; Smeekens, J.; Wu, R. Evaluation and optimization of reduction and alkylation methods to maximize peptide identification with MS-based proteomics. Mol. BioSyst. 2017, 13, 2574–2582. [Google Scholar] [CrossRef] [PubMed]
  55. Müller, T.; Winter, D. Systematic evaluation of protein reduction and alkylation reveals massive unspecific side effects by iodine-containing reagents. Mol. Cell. Proteom. 2017, 16, 1173–1187. [Google Scholar] [CrossRef] [PubMed]
  56. Kuznetsova, K.G.; Levitsky, L.I.; Pyatnitskiy, M.A.; Ilina, I.Y.; Bubis, J.A.; Solovyeva, E.M.; Zgoda, V.G.; Gorshkov, M.V.; Moshkovskii, S.A. Cysteine alkylation methods in shotgun proteomics and their possible effects on methionine residues. J. Proteom. 2021, 231, 104022. [Google Scholar] [CrossRef]
  57. Hains, P.G.; Robinson, P.J. The impact of commonly used alkylating agents on artifactual peptide modification. J. Proteome Res. 2017, 16, 3443–3447. [Google Scholar] [CrossRef]
  58. Geoghegan, K.F.; Hoth, L.R.; Tan, D.H.; Borzilleri, K.A.; Withka, J.M.; Boyd, J.G. Cyclization of N-terminal S-carbamoylmethylcysteine causing loss of 17 Da from peptides and extra peaks in peptide maps. J. Proteome Res. 2002, 1, 181–187. [Google Scholar] [CrossRef]
  59. Switzar, L.; Giera, M.; Niessen, W.M. Protein digestion: An overview of the available techniques and recent developments. J. Proteome Res. 2013, 12, 1067–1077. [Google Scholar] [CrossRef]
  60. Falck, D.; Jansen, B.C.; Plomp, R.; Reusch, D.; Haberger, M.; Wuhrer, M. Glycoforms of immunoglobulin G based biopharmaceuticals are differentially cleaved by trypsin due to the glycoform influence on higher-order structure. J. Proteome Res. 2015, 14, 4019–4028. [Google Scholar] [CrossRef]
  61. Solari, F.A.; Dell’Aica, M.; Sickmann, A.; Zahedi, R.P. Why phosphoproteomics is still a challenge. Mol. BioSystems 2015, 11, 1487–1493. [Google Scholar] [CrossRef]
  62. Heissel, S.; Frederiksen, S.J.; Bunkenborg, J.; Højrup, P. Enhanced trypsin on a budget: Stabilization, purification and high-temperature application of inexpensive commercial trypsin for proteomics applications. PLoS ONE 2019, 14, e0218374. [Google Scholar] [CrossRef] [PubMed]
  63. Menneteau, T.; Saveliev, S.; Butré, C.I.; Rivera, A.K.G.; Urh, M.; Delobel, A. Addressing common challenges of biotherapeutic protein peptide mapping using recombinant trypsin. J. Pharm. Biomed. Anal. 2024, 243, 116124. [Google Scholar] [CrossRef] [PubMed]
  64. Shibata, K.; Naito, T.; Okamura, J.; Hosokawa, S.; Mineta, H.; Kawakami, J. Simple and rapid LC-MS/MS method for the absolute determination of cetuximab in human serum using an immobilized trypsin. J. Pharm. Biomed. Anal. 2017, 146, 266–272. [Google Scholar] [CrossRef] [PubMed]
  65. Reinders, L.M.; Klassen, M.D.; Teutenberg, T.; Jaeger, M.; Schmidt, T.C. Development of a multidimensional online method for the characterization and quantification of monoclonal antibodies using immobilized flow-through enzyme reactors. Anal. Bioanal. Chem. 2021, 413, 7119–7128. [Google Scholar] [CrossRef]
  66. Nagy, C.; Szabo, R.; Gaspar, A. Microfluidic immobilized enzymatic reactors for proteomic analyses—Recent developments and trends (2017–2021). Micromachines 2022, 13, 311. [Google Scholar] [CrossRef]
  67. Glatter, T.; Ludwig, C.; Ahrné, E.; Aebersold, R.; Heck, A.J.R.; Schmidt, A. Large-Scale Quantitative Assessment of Different In-Solution Protein Digestion Protocols Reveals Superior Cleavage Efficiency of Tandem Lys-C/Trypsin Proteolysis over Trypsin Digestion. J. Proteome Res. 2012, 11, 5145–5156. [Google Scholar] [CrossRef]
  68. Giansanti, P.; Tsiatsiani, L.; Low, T.Y.; Heck, A.J.R. Six alternative proteases for mass spectrometry–based proteomics beyond trypsin. Nat. Protoc. 2016, 11, 993–1006. [Google Scholar] [CrossRef]
  69. Trevisiol, S.; Ayoub, D.; Lesur, A.; Ancheva, L.; Gallien, S.; Domon, B. The use of proteases complementary to trypsin to probe isoforms and modifications. Proteomics 2016, 16, 715–728. [Google Scholar] [CrossRef]
  70. Hao, P.; Ren, Y.; Datta, A.; Tam, J.P.; Sze, S.K. Evaluation of the effect of trypsin digestion buffers on artificial deamidation. J. Proteome Res. 2015, 14, 1308–1314. [Google Scholar] [CrossRef]
  71. Sutherland, E.; Veth, T.S.; Riley, N.M. Revisiting the effect of trypsin digestion buffers on artificial deamidation. ChemRxiv 2024. [Google Scholar] [CrossRef]
  72. Jiang, H.; Zeng, J.; Titsch, C.; Voronin, K.; Akinsanya, B.; Luo, L.; Shen, H.; Desai, D.D.; Allentoff, A.; Aubry, A.-F.; et al. Fully Validated LC-MS/MS Assay for the Simultaneous Quantitation of Coadministered Therapeutic Antibodies in Cynomolgus Monkey Serum. Anal. Chem. 2013, 85, 9859–9867. [Google Scholar] [CrossRef] [PubMed]
  73. Ren, D.; Pipes, G.D.; Liu, D.; Shih, L.-Y.; Nichols, A.C.; Treuheit, M.J.; Brems, D.N.; Bondarenko, P.V. An improved trypsin digestion method minimizes digestion-induced modifications on proteins. Anal. Biochem. 2009, 392, 12–21. [Google Scholar] [CrossRef] [PubMed]
  74. Liu, H.; Ponniah, G.; Neill, A.; Patel, R.; Andrien, B. Accurate determination of protein methionine oxidation by stable isotope labeling and LC-MS analysis. Anal. Chem. 2013, 85, 11705–11709. [Google Scholar] [CrossRef]
  75. Yang, J.; Gao, Z.; Ren, X.; Sheng, J.; Xu, P.; Chang, C.; Fu, Y. DeepDigest: Prediction of Protein Proteolytic Digestion with Deep Learning. Anal. Chem. 2021, 93, 6094–6103. [Google Scholar] [CrossRef]
  76. Gupta, S.; Jiskoot, W.; Schöneich, C.; Rathore, A.S. Oxidation and deamidation of monoclonal antibody products: Potential impact on stability, biological activity, and efficacy. J. Pharm. Sci. 2022, 111, 903–918. [Google Scholar] [CrossRef]
  77. Millán-Martín, S.; Jakes, C.; Carillo, S.; Buchanan, T.; Guender, M.; Kristensen, D.B.; Sloth, T.M.; Ørgaard, M.; Cook, K.; Bones, J. Inter-laboratory study of an optimised peptide mapping workflow using automated trypsin digestion for monitoring monoclonal antibody product quality attributes. Anal. Bioanal. Chem. 2020, 412, 6833–6848. [Google Scholar] [CrossRef]
  78. Kuzyk, M.A.; Parker, C.E.; Domanski, D.; Borchers, C.H. Development of MRM-based assays for the absolute quantitation of plasma proteins. Methods Mol. Biol. 2013, 1023, 53–82. [Google Scholar] [CrossRef]
  79. Liebler, D.C.; Zimmerman, L.J. Targeted quantitation of proteins by mass spectrometry. Biochemistry 2013, 52, 3797–3806. [Google Scholar] [CrossRef]
  80. Schmidt, C.; Urlaub, H. Absolute quantification of proteins using standard peptides and multiple reaction monitoring. Quant. Methods Proteom. 2012, 893, 249–265. [Google Scholar] [CrossRef]
  81. Gupta, R.; Brunak, S. Prediction of glycosylation across the human proteome and the correlation to protein function. Pac Symp Biocomput. 2002, 310–322. [Google Scholar] [CrossRef]
  82. Pugalenthi, G.; Nithya, V.; Chou, K.-C.; Archunan, G. Nglyc: A random forest method for prediction of N-glycosylation sites in eukaryotic protein sequence. Protein Pept. Lett. 2020, 27, 178–186. [Google Scholar] [CrossRef] [PubMed]
  83. Pakhrin, S.C.; Aoki-Kinoshita, K.F.; Caragea, D.; Kc, D.B. DeepNGlyPred: A deep neural network-based approach for human N-linked glycosylation site prediction. Molecules 2021, 26, 7314. [Google Scholar] [CrossRef] [PubMed]
  84. Jian, W.; Kang, L.; Burton, L.; Weng, N. A workflow for absolute quantitation of large therapeutic proteins in biological samples at intact level using LC-HRMS. Bioanalysis 2016, 8, 1679–1691. [Google Scholar] [CrossRef] [PubMed]
  85. Lanshoeft, C.; Cianférani, S.; Heudi, O. Generic Hybrid Ligand Binding Assay Liquid Chromatography High-Resolution Mass Spectrometry-Based Workflow for Multiplexed Human Immunoglobulin G1 Quantification at the Intact Protein Level: Application to Preclinical Pharmacokinetic Studies. Anal. Chem. 2017, 89, 2628–2635. [Google Scholar] [CrossRef]
  86. Cong, Y.; Zhang, Z.; Zhang, S.; Hu, L.; Gu, J. Quantitative MS analysis of therapeutic mAbs and their glycosylation for pharmacokinetics study. Proteom.—Clin. Appl. 2016, 10, 303–314. [Google Scholar] [CrossRef]
  87. Liu, S.; Liu, X. IgG N-glycans. Adv. Clin. Chem. 2021, 105, 1–47. [Google Scholar] [CrossRef]
  88. van den Broek, I.; van Dongen, W.D. LC–MS-based quantification of intact proteins: Perspective for clinical and bioanalytical applications. Bioanalysis 2015, 7, 1943–1958. [Google Scholar] [CrossRef]
  89. Rosati, S.; Yang, Y.; Barendregt, A.; Heck, A.J.R. Detailed mass analysis of structural heterogeneity in monoclonal antibodies using native mass spectrometry. Nat. Protoc. 2014, 9, 967–976. [Google Scholar] [CrossRef]
  90. Xu, K.; Liu, L.; Saad, O.M.; Baudys, J.; Williams, L.; Leipold, D.; Shen, B.; Raab, H.; Junutula, J.R.; Kim, A. Characterization of intact antibody–drug conjugates from plasma/serum in vivo by affinity capture capillary liquid chromatography–mass spectrometry. Anal. Biochem. 2011, 412, 56–66. [Google Scholar] [CrossRef]
  91. Jin, W.; Burton, L.; Moore, I. LC–HRMS quantitation of intact antibody drug conjugate trastuzumab emtansine from rat plasma. Bioanalysis 2018, 10, 851–862. [Google Scholar] [CrossRef]
  92. Zhao, Y.; Gu, H.; Zheng, N.; Zeng, J. Critical considerations for immunocapture enrichment LC–MS bioanalysis of protein therapeutics and biomarkers. Bioanalysis 2018, 10, 987–995. [Google Scholar] [CrossRef] [PubMed]
  93. Kang, L.; Weng, N.; Jian, W. LC–MS bioanalysis of intact proteins and peptides. Biomed. Chromatogr. 2020, 34, e4633. [Google Scholar] [CrossRef] [PubMed]
  94. Zhang, L.; Vasicek, L.A.; Hsieh, S.; Zhang, S.; Bateman, K.P.; Henion, J. Top-down LC–MS quantitation of intact denatured and native monoclonal antibodies in biological samples. Bioanalysis 2018, 10, 1039–1054. [Google Scholar] [CrossRef] [PubMed]
  95. Song, Y.E.; Lebert, D.; Guillaubez, J.-V.; Samra, S.; Goucher, E.; Hart, B. Streamlined Workflow for Absolute Quantitation of Therapeutic Monoclonal Antibodies Using Promise Proteomics mAbXmise Kits and a TSQ Altis Plus Mass Spectrometer. Available online: https://promise-proteomics.com/wp-content/uploads/2023/06/tn-001753-cl-clinical-altis-mabs-tn001753-na-en.pdf (accessed on 7 August 2024).
  96. Rutherfurd, S.M.; Gilani, G.S. Amino Acid Analysis. Curr. Protoc. Protein Sci. 2009, 58, 11.9.1–11.9.37. [Google Scholar] [CrossRef]
  97. Rutherfurd, S.M.; Dunn, B.M. Quantitative Amino Acid Analysis. Curr. Protoc. Protein Sci. 2011, 63, 3.2.1–3.2.6. [Google Scholar] [CrossRef]
  98. Josephs, R.D.; Martos, G.; Li, M.; Wu, L.; Melanson, J.E.; Quaglia, M.; Beltrão, P.J.; Prevoo-Franzsen, D.; Boeuf, A.; Delatour, V. Establishment of measurement traceability for peptide and protein quantification through rigorous purity assessment—A review. Metrologia 2019, 56, 044006. [Google Scholar] [CrossRef]
  99. Violi, J.P.; Bishop, D.P.; Padula, M.P.; Steele, J.R.; Rodgers, K.J. Considerations for amino acid analysis by liquid chromatography-tandem mass spectrometry: A tutorial review. TrAC Trends Anal. Chem. 2020, 131, 116018. [Google Scholar] [CrossRef]
  100. Liu, F.; Lai, S.; Tong, H.; Lakey, P.S.; Shiraiwa, M.; Weller, M.G.; Pöschl, U.; Kampf, C.J. Release of free amino acids upon oxidation of peptides and proteins by hydroxyl radicals. Anal. Bioanal. Chem. 2017, 409, 2411–2420. [Google Scholar] [CrossRef]
  101. Kato, M.; Kato, H.; Eyama, S.; Takatsu, A. Application of amino acid analysis using hydrophilic interaction liquid chromatography coupled with isotope dilution mass spectrometry for peptide and protein quantification. J. Chromatogr. B 2009, 877, 3059–3064. [Google Scholar] [CrossRef]
  102. Stocks, B.B.; Thibeault, M.-P.; Schrag, J.D.; Melanson, J.E. Characterization of a SARS-CoV-2 spike protein reference material. Anal. Bioanal. Chem. 2022, 414, 3561–3569. [Google Scholar] [CrossRef]
  103. Stocks, B.B.; Thibeault, M.-P.; L’Abbé, D.; Stuible, M.; Durocher, Y.; Melanson, J.E. Production and Characterization of a SARS-CoV-2 Nucleocapsid Protein Reference Material. ACS Meas. Sci. 2022, 2, 620–628. [Google Scholar] [CrossRef] [PubMed]
  104. Louwagie, M.; Kieffer-Jaquinod, S.; Dupierris, V.; Couté, Y.; Bruley, C.; Garin, J.; Dupuis, A.; Jaquinod, M.; Brun, V. Introducing AAA-MS, a Rapid and Sensitive Method for Amino Acid Analysis Using Isotope Dilution and High-Resolution Mass Spectrometry. J. Proteome Res. 2012, 11, 3929–3936. [Google Scholar] [CrossRef] [PubMed]
  105. Jeong, J.-S.; Lim, H.-M.; Kim, S.-K.; Ku, H.-K.; Oh, K.-H.; Park, S.-R. Quantification of human growth hormone by amino acid composition analysis using isotope dilution liquid-chromatography tandem mass spectrometry. J. Chromatogr. A 2011, 1218, 6596–6602. [Google Scholar] [CrossRef] [PubMed]
  106. Mi, W.; Josephs, R.; Melanson, J.; Dai, X.; Wang, Y.; Zhai, R.; Chu, Z.; Fang, X.; Thibeault, M.; Stocks, B. PAWG Pilot Study on Quantification of SARS-CoV-2 Monoclonal Antibody—Part 1. Metrologia 2022, 59, 08001. [Google Scholar] [CrossRef]
  107. Stocks, B.B.; Thibeault, M.-P.; L’Abbé, D.; Umer, M.; Liu, Y.; Stuible, M.; Durocher, Y.; Melanson, J.E. Characterization of biotinylated human ACE2 and SARS-CoV-2 Omicron BA. 4/5 spike protein reference materials. Anal. Bioanal. Chem. 2024, 416, 4861–4872. [Google Scholar] [CrossRef]
  108. Catalogue: SIL-mAbs for Targeted LC-MS Quantification. Available online: https://promise-proteomics.com/wp-content/uploads/2024/01/Catalogue-SIL-mAbs-2024.pdf (accessed on 7 August 2024).
  109. Picard, G.; Lebert, D.; Louwagie, M.; Adrait, A.; Huillet, C.; Vandenesch, F.; Bruley, C.; Garin, J.; Jaquinod, M.; Brun, V. PSAQ™ standards for accurate MS-based quantification of proteins: From the concept to biomedical applications. J. Mass Spectrom. 2012, 47, 1353–1363. [Google Scholar] [CrossRef]
  110. Ong, S.-E.; Blagoev, B.; Kratchmarova, I.; Kristensen, D.B.; Steen, H.; Pandey, A.; Mann, M. Stable Isotope Labeling by Amino Acids in Cell Culture, SILAC, as a Simple and Accurate Approach to Expression Proteomics. Mol. Cell. Proteom. 2002, 1, 376–386. [Google Scholar] [CrossRef]
  111. Osaki, F.; Tabata, K.; Oe, T. Quantitative LC/ESI-SRM/MS of antibody biopharmaceuticals: Use of a homologous antibody as an internal standard and three-step method development. Anal. Bioanal. Chem. 2017, 409, 5523–5532. [Google Scholar] [CrossRef]
  112. Smit, N.P.M.; Ruhaak, L.R.; Romijn, F.P.H.T.M.; Pieterse, M.M.; Van Der Burgt, Y.E.M.; Cobbaert, C.M. The Time Has Come for Quantitative Protein Mass Spectrometry Tests That Target Unmet Clinical Needs. J. Am. Soc. Mass Spectrom. 2021, 32, 636–647. [Google Scholar] [CrossRef]
  113. Lee, H.; Lee, J. Peptide purity assignment for antibody quantification by combining isotope dilution mass spectrometry and liquid chromatography. Bull. Korean Chem. Soc. 2022, 43, 704–713. [Google Scholar] [CrossRef]
  114. Burkitt, W.I.; Pritchard, C.; Arsene, C.; Henrion, A.; Bunk, D.; O’Connor, G. Toward Systeme International d’Unite-traceable protein quantification: From amino acids to proteins. Anal. Biochem. 2008, 376, 242–251. [Google Scholar] [CrossRef] [PubMed]
  115. Benesova, E.; Vidova, V.; Spacil, Z. A comparative study of synthetic winged peptides for absolute protein quantification. Sci. Rep. 2021, 11, 10880. [Google Scholar] [CrossRef] [PubMed]
  116. Li, H.; Ortiz, R.; Tran, L.; Hall, M.; Spahr, C.; Walker, K.; Laudemann, J.; Miller, S.; Salimi-Moosavi, H.; Lee, J.W. General LC-MS/MS Method Approach to Quantify Therapeutic Monoclonal Antibodies Using a Common Whole Antibody Internal Standard with Application to Preclinical Studies. Anal. Chem. 2012, 84, 1267–1273. [Google Scholar] [CrossRef]
  117. Bronsema, K.J.; Bischoff, R.; van de Merbel, N.C. Internal standards in the quantitative determination of protein biopharmaceuticals using liquid chromatography coupled to mass spectrometry. J. Chromatogr. B 2012, 893, 1–14. [Google Scholar] [CrossRef]
  118. Furlong, M.T.; Ouyang, Z.; Wu, S.; Tamura, J.; Olah, T.; Tymiak, A.; Jemal, M. A universal surrogate peptide to enable LC-MS/MS bioanalysis of a diversity of human monoclonal antibody and human Fc-fusion protein drug candidates in pre-clinical animal studies. Biomed. Chromatogr. 2012, 26, 1024–1032. [Google Scholar] [CrossRef]
  119. Furlong, M.T. Generic Peptide Strategies for LC–MS/MS Bioanalysis of Human Monoclonal Antibody Drugs and Drug Candidates. In Protein Analysis Using Mass Spectrometry: Accelerating Protein Biotherapeutics from Lab to Patient; John Wiley & Sons: Hoboken, NJ, USA, 2017; pp. 161–181. [Google Scholar]
  120. Food and Drug Administration. Bioanalytical Method Validation Guidance for Industry. Available online: https://www.fda.gov/regulatory-information/search-fda-guidance-documents/bioanalytical-method-validation-guidance-industry (accessed on 7 August 2024).
  121. European Medicines Agency. ICH Guideline M10 on Bioanalytical Method Validation and Study Sample Analysis. Available online: https://www.ema.europa.eu/en/documents/scientific-guideline/ich-guideline-m10-bioanalytical-method-validation-step-5_en.pdf (accessed on 7 August 2024).
  122. Vidova, V.; Spacil, Z. A review on mass spectrometry-based quantitative proteomics: Targeted and data independent acquisition. Anal. Chim. Acta 2017, 964, 7–23. [Google Scholar] [CrossRef]
  123. Ranbaduge, N.; Yu, Y.Q. A Streamlined Compliant Ready Workflow for Peptide-Based Multi-Attribute Method (MAM). Available online: https://www.waters.com/content/dam/waters/en/app-notes/2020/720007094/720007094-zh_tw.pdf (accessed on 7 August 2024).
  124. Yun, W.; Alelyunas, H.S.; Wrona, M.D.; Chen, W. Rapid, Sensitive, and Routine Intact mAb Quantification Using a Compact Tof HRMS Platform. Available online: https://lcms.cz/labrulez-bucket-strapi-h3hsga3/2019asms_alelyunas_intactproteinquan_659b6f83aa/2019asms_alelyunas_intactproteinquan.pdf (accessed on 7 August 2024).
  125. Millán-Martín, S.; Jakes, C.; Carillo, S.; Bones, J. Multi-Attribute Method (MAM) Analytical Workflow for Biotherapeutic Protein Characterization from Process Development to QC. Curr. Protoc. 2023, 3, e927. [Google Scholar] [CrossRef]
  126. Schneck, N.A.; Mehl, J.T.; Kellie, J.F. Protein LC-MS Tools for the Next Generation of Biotherapeutic Analyses from Preclinical and Clinical Serum. J. Am. Soc. Mass Spectrom. 2023, 34, 1837–1846. [Google Scholar] [CrossRef]
  127. Kiyonami, R.; Schoen, A.; Prakash, A.; Nguyen, H.; Peterman, S.; Selevsek, N.; Zabrouskov, V.; Huhmer, A.; Domon, B. Rapid Assay Development and Refinement for Targeted Protein Quantitation Using an Intelligent SRM (iSRM) Workflow. Available online: https://tools.thermofisher.com/content/sfs/brochures/AN468_63139_Vantage_Prot(1).pdf (accessed on 7 August 2024).
  128. Kiyonami, R.; Zeller, M.; Zabrouskov, V. Quantifying Peptides in Complex Mixtures with High Sensitivity and Precision Using a Targeted Approach with a Hybrid Linear Ion Trap-Orbitrap Mass Spectrometer. Available online: https://assets.thermofisher.com/TFS-Assets/CMD/Application-Notes/AN-557-LC-MS-Peptides-Complex-Mixtures-AN63499-EN.pdf (accessed on 7 August 2024).
  129. Colangelo, C.M.; Chung, L.; Bruce, C.; Cheung, K.-H. Review of software tools for design and analysis of large scale MRM proteomic datasets. Methods 2013, 61, 287–298. [Google Scholar] [CrossRef]
  130. Desiere, F.; Deutsch, E.W.; King, N.L.; Nesvizhskii, A.I.; Mallick, P.; Eng, J.; Chen, S.; Eddes, J.; Loevenich, S.N.; Aebersold, R. The PeptideAtlas project. Nucleic Acids Res. 2006, 34, D655–D658. [Google Scholar] [CrossRef]
  131. MacLean, B.; Tomazela, D.M.; Shulman, N.; Chambers, M.; Finney, G.L.; Frewen, B.; Kern, R.; Tabb, D.L.; Liebler, D.C.; MacCoss, M.J. Skyline: An open source document editor for creating and analyzing targeted proteomics experiments. Bioinformatics 2010, 26, 966–968. [Google Scholar] [CrossRef] [PubMed]
  132. Gessulat, S.; Schmidt, T.; Zolg, D.P.; Samaras, P.; Schnatbaum, K.; Zerweck, J.; Knaute, T.; Rechenberger, J.; Delanghe, B.; Huhmer, A. Prosit: Proteome-wide prediction of peptide tandem mass spectra by deep learning. Nat. Methods 2019, 16, 509–518. [Google Scholar] [CrossRef] [PubMed]
  133. Cox, J.; Mann, M. MaxQuant enables high peptide identification rates, individualized ppb-range mass accuracies and proteome-wide protein quantification. Nat. Biotechnol. 2008, 26, 1367–1372. [Google Scholar] [CrossRef]
  134. Cox, J.; Matic, I.; Hilger, M.; Nagaraj, N.; Selbach, M.; Olsen, J.V.; Mann, M. A practical guide to the MaxQuant computational platform for SILAC-based quantitative proteomics. Nat. Protoc. 2009, 4, 698–705. [Google Scholar] [CrossRef] [PubMed]
  135. Tyanova, S.; Temu, T.; Cox, J. The MaxQuant computational platform for mass spectrometry-based shotgun proteomics. Nat. Protoc. 2016, 11, 2301–2319. [Google Scholar] [CrossRef] [PubMed]
  136. Röst, H.L.; Sachsenberg, T.; Aiche, S.; Bielow, C.; Weisser, H.; Aicheler, F.; Andreotti, S.; Ehrlich, H.-C.; Gutenbrunner, P.; Kenar, E. OpenMS: A flexible open-source software platform for mass spectrometry data analysis. Nat. Methods 2016, 13, 741–748. [Google Scholar] [CrossRef]
  137. Pfeuffer, J.; Bielow, C.; Wein, S.; Jeong, K.; Netz, E.; Walter, A.; Alka, O.; Nilse, L.; Colaianni, P.D.; McCloskey, D. OpenMS 3 enables reproducible analysis of large-scale mass spectrometry data. Nat. Methods 2024, 21, 365–367. [Google Scholar] [CrossRef]
  138. Tsugawa, H.; Kanazawa, M.; Ogiwara, A.; Arita, M. MRMPROBS suite for metabolomics using large-scale MRM assays. Bioinformatics 2014, 30, 2379–2380. [Google Scholar] [CrossRef]
  139. Valot, B.; Langella, O.; Nano, E.; Zivy, M. MassChroQ: A versatile tool for mass spectrometry quantification. Proteomics 2011, 11, 3572–3577. [Google Scholar] [CrossRef]
  140. Cai, Y.; Weng, K.; Guo, Y.; Peng, J.; Zhu, Z.-J. An integrated targeted metabolomic platform for high-throughput metabolite profiling and automated data processing. Metabolomics 2015, 11, 1575–1586. [Google Scholar] [CrossRef]
  141. R Core Team. R: A Language and Environment for Statistical Computing; R Foundation for Statistical Computing: Vienna, Austria, 2022; Available online: https://www.R-project.org/ (accessed on 7 August 2024).
  142. Choi, M.; Carver, J.; Chiva, C.; Tzouros, M.; Huang, T.; Tsai, T.-H.; Pullman, B.; Bernhardt, O.M.; Hüttenhain, R.; Teo, G.C. MassIVE. quant: A community resource of quantitative mass spectrometry–based proteomics datasets. Nat. Methods 2020, 17, 981–984. [Google Scholar] [CrossRef] [PubMed]
  143. Dai, C.; Pfeuffer, J.; Wang, H.; Zheng, P.; Käll, L.; Sachsenberg, T.; Demichev, V.; Bai, M.; Kohlbacher, O.; Perez-Riverol, Y. quantms: A cloud-based pipeline for quantitative proteomics enables the reanalysis of public proteomics data. Nat. Methods 2024, 21, 1603–1607. [Google Scholar] [CrossRef] [PubMed]
  144. Wang, R.; Jiang, H.; Lu, M.; Tong, J.; An, S.; Wang, J.; Yu, C. MRMPro: A web-based tool to improve the speed of manual calibration for multiple reaction monitoring data analysis by mass spectrometry. BMC Bioinform. 2024, 25, 60. [Google Scholar] [CrossRef]
  145. Mann, M.; Kumar, C.; Zeng, W.-F.; Strauss, M.T. Artificial intelligence for proteomics and biomarker discovery. Cell Syst. 2021, 12, 759–770. [Google Scholar] [CrossRef]
  146. Mi, W.; Josephs, R.; Melanson, J.; Dai, X.; Wang, Y.; Zhai, R.; Chu, Z.; Fang, X.; Thibeault, M.; Stocks, B. PAWG pilot study on quantification of SARS-CoV-2 monoclonal antibody—Part 2. Metrologia 2023, 60, 08016. [Google Scholar] [CrossRef]
  147. Miller, W.G.; Greenberg, N.; Panteghini, M.; Budd, J.R.; Johansen, J.V. Guidance on which calibrators in a metrologically traceable calibration hierarchy must be commutable with clinical samples. Clin. Chem. 2023, 69, 228–238. [Google Scholar] [CrossRef]
  148. Diederiks, N.M.; van der Burgt, Y.E.; Ruhaak, L.R.; Cobbaert, C.M. Developing an SI-traceable Lp (a) reference measurement system: A pilgrimage to selective and accurate apo (a) quantification. Crit. Rev. Clin. Lab. Sci. 2023, 60, 483–501. [Google Scholar] [CrossRef]
  149. Ruhaak, L.R.; Romijn, F.P.; Begcevic Brkovic, I.; Kuklenyik, Z.; Dittrich, J.; Ceglarek, U.; Hoofnagle, A.N.; Althaus, H.; Angles-Cano, E.; Coassin, S. Development of an LC-MRM-MS-based candidate reference measurement procedure for standardization of serum apolipoprotein (a) tests. Clin. Chem. 2023, 69, 251–261. [Google Scholar] [CrossRef]
  150. Schiel, J.E.; Mire-Sluis, A.; Davis, D. Monoclonal antibody therapeutics: The need for biopharmaceutical reference materials. In State-of-the-Art and Emerging Technologies for Therapeutic Monoclonal Antibody Characterization Volume 1. Monoclonal Antibody Therapeutics: Structure, Function, and Regulatory Space; ACS Publications: Washington, DC, USA, 2014; pp. 1–34. [Google Scholar]
  151. Schiel, J.E.; Turner, A. The NISTmAb Reference Material 8671 lifecycle management and quality plan. Anal. Bioanal. Chem. 2018, 410, 2067–2078. [Google Scholar] [CrossRef]
  152. Kinumi, T.; Saikusa, K.; Kato, M.; Kojima, R.; Igarashi, C.; Noda, N.; Honda, S. Characterization and Value Assignment of a Monoclonal Antibody Reference Material, NMIJ RM 6208a, AIST-MAB. Front. Mol. Biosci. 2022, 9, 842041. [Google Scholar] [CrossRef]
  153. Dong, L.; Zhang, Y.; Fu, B.; Swart, C.; Jiang, H.; Liu, Y.; Huggett, J.; Wielgosz, R.; Niu, C.; Li, Q. Reliable biological and multi-omics research through biometrology. Anal. Bioanal. Chem. 2024, 416, 3645–3663. [Google Scholar] [CrossRef] [PubMed]
  154. Shuford, C.M.; Walters, J.J.; Holland, P.M.; Sreenivasan, U.; Askari, N.; Ray, K.; Grant, R.P. Absolute protein quantification by mass spectrometry: Not as simple as advertised. Anal. Chem. 2017, 89, 7406–7415. [Google Scholar] [CrossRef] [PubMed]
  155. Zhong, X.; Chen, H.; Zare, R.N. Ultrafast enzymatic digestion of proteins by microdroplet mass spectrometry. Nat. Commun. 2020, 11, 1049. [Google Scholar] [CrossRef] [PubMed]
  156. Xiao, T.; Li, Z.; Xing, X.; He, F.; Huang, J.; Xue, D. Improving the activity and thermal stability of trypsin by the rational design. Process Biochem. 2023, 130, 227–235. [Google Scholar] [CrossRef]
  157. Guo, C.; Liu, Y.; Yu, H.; Du, K.; Gan, Y.; Huang, H. A novel strategy for thermostability improvement of trypsin based on N-glycosylation within the Ω-loop region. J. Microbiol. Biotechnol. 2016, 26, 1163–1172. [Google Scholar] [CrossRef]
  158. Narumi, R.; Masuda, K.; Tomonaga, T.; Adachi, J.; Ueda, H.R.; Shimizu, Y. Cell-free synthesis of stable isotope-labeled internal standards for targeted quantitative proteomics. Synth. Syst. Biotechnol. 2018, 3, 97–104. [Google Scholar] [CrossRef]
Figure 2. Overview of MS-based antibody quantification strategies of pure or matrix-containing antibody samples. Proteolytic enzymes cleave the antibody sequence at specific positions, such as the C-terminal bond of lysine and arginine (trypsin), releasing defined peptide fragments. The treatment of antibodies with acids under high temperatures enables quantification of free amino acids.
Figure 2. Overview of MS-based antibody quantification strategies of pure or matrix-containing antibody samples. Proteolytic enzymes cleave the antibody sequence at specific positions, such as the C-terminal bond of lysine and arginine (trypsin), releasing defined peptide fragments. The treatment of antibodies with acids under high temperatures enables quantification of free amino acids.
Antibodies 14 00003 g002
Figure 3. Cleavage sites of commonly used proteases for bottom-up proteomics.
Figure 3. Cleavage sites of commonly used proteases for bottom-up proteomics.
Antibodies 14 00003 g003
Figure 4. Schematic principle of SIL internal standard integration for the different quantification methods of mAbs. The sample is spiked with known concentration of SIL internal standard antibody, peptide, or amino acids (red). Due to the chemical similarity, both sample and SIL standard elute at the same time and differ only in their mass-to-charge (m/z) ratios.
Figure 4. Schematic principle of SIL internal standard integration for the different quantification methods of mAbs. The sample is spiked with known concentration of SIL internal standard antibody, peptide, or amino acids (red). Due to the chemical similarity, both sample and SIL standard elute at the same time and differ only in their mass-to-charge (m/z) ratios.
Antibodies 14 00003 g004
Table 1. Top 5 best-selling recombinant antibodies approved for therapeutic application. 1 Yearly turnover in 2023 [21]. 2 Published peptide-based quantification methods within the last 5 years for the respective mAb and the selected signature peptides.
Table 1. Top 5 best-selling recombinant antibodies approved for therapeutic application. 1 Yearly turnover in 2023 [21]. 2 Published peptide-based quantification methods within the last 5 years for the respective mAb and the selected signature peptides.
Generic NameAntibody SubclassYear of
Approval
TargetSales 1
(Billion USD)
Signature Peptides 2
PembrolizumabIgG4κ2021Programmed cell death protein 1 (PD-1)25.0ASGYTFTNYYMYWVR [28]
DLPLTFGGGTK [28,29]
VTLTTDSSTTTAYMELK [30]
AdalimumabIgG1κ2002Tumor necrosis
factor alpha (TNF-α)
14.4APYTFGQGTK [31,32]
UstekinumabIgG1κ2009Interleukin (IL)-12 and IL-2310.9PGQGYFDFWGQGTLVTVSSSSTK [33]
GLDWIGIMSPVDSDIR [29,33]
DaratumumabIgG1κ2016Hydrolase CD38 9.7SNWPPTFGQGTK [34]
LLIYDASNR [35]
NivolumabIgG4κ2014PD-19.0ASGITFSNSGMHWVR [29,30,36,37]
Table 2. Exemplary overview of commercially available MS-grade trypsin and trypsin/Lys-C variants, together with their recommended sample preparation procedures for HPLC-MS/MS analysis and reference to their use for quantification of therapeutic antibodies from Table 1. Products from other manufacturers with similar properties may be equivalent. DTT: Dithiothreitol; TCEP: Tris(2-carboxyethyl)phosphine; IAA: Iodoacetic acid, IAM: Iodoacetamide; opt.: optional.
Table 2. Exemplary overview of commercially available MS-grade trypsin and trypsin/Lys-C variants, together with their recommended sample preparation procedures for HPLC-MS/MS analysis and reference to their use for quantification of therapeutic antibodies from Table 1. Products from other manufacturers with similar properties may be equivalent. DTT: Dithiothreitol; TCEP: Tris(2-carboxyethyl)phosphine; IAA: Iodoacetic acid, IAM: Iodoacetamide; opt.: optional.
Product Special
Feature
DenaturationReductionAlkylationDigestion
Conditions
Ref.
Trypsin
Promega GmbH
(Walldorf, BW, GER)
Trypsin Gold/Sequencing GradeMaximum specificity8 M Urea
1 h
DTTIAM
30 min
Overnight
37 °C
[28,31,37]
Trypsin PlatinumRecombinant enzyme,
autoproteolytic resistance
8 M GuHCl
30 min
TCEPIAM
30 min
Overnight
37 °C
Thermo Fisher Scientific
(Waltham, MA, USA)
Pierce™ Trypsin 1 h at 60 °C or
10 min at 95 °C
DTTIAA,
30 min
4 to 24 h
37 °C
[29,30]
SMART Digest Trypsin-KitAutomatable process-opt.opt.45 min (IgG)
70 °C
In-Solution Tryptic Digestion and Guanidination KitImproved
ionization by guanidination of K into homo-R
95 °C
5 min
DTTIAM,
30 min
2 h at 37 °C or overnight
at 30 °C
Waters Corporation
(Milford, MA, USA)
ProteinWorks
eXpress Digest Kit
High throughput of samples possibleDigestion buffer, 80 °C, 10 minReduction agent
60 °C, 20 min
Alkylation
agent
30 min
2 h
45 °C
[33]
Promise Proteomics
(Grenoble, ARA, FRA)
mAbXmise KitImmunocapture
cartridges
opt.,
4 M to 0.1 M Urea
--30 min to 15 h
37 °C
[32]
Trypsin/ Lys-C Mix
Thermo Fisher Scientific
(Waltham, MA, USA)
EasyPep™ Mini
MS Sample Prep Kit
High throughput of samples possibleLysis solution
95 °C, 10 min
Red.
Solution
Alk.
Solution
1 to 3 h
37 °C
Pierce™ Trypsin/ Lys-C Protease Mix 8 M Urea,
1 h at 60 °C or
10 min at 95 °C
DTTIAM,
30 min
2 to 16 h
37 °C
Promega GmbH
(Walldorf, BW, GER)
Rapid Digestion–
Trypsin/LysC
Fast digestion-opt.opt.1 h
70 °C
Trypsin/Lys-CQuantification6–8 M Urea,
30 min
DTTIAM,
30 min
overnight
37 °C
Table 3. Brief overview of frequently used software including latest version for development and evaluation of targeted mAb quantification experiments.1 Open-source software. 2 Specifically designed for HRMS data.
Table 3. Brief overview of frequently used software including latest version for development and evaluation of targeted mAb quantification experiments.1 Open-source software. 2 Specifically designed for HRMS data.
Manufacturer/LabSoftware
Waters Coporation
(Milford, MA, USA)
BioAccord System (UNIFI software, version 1.9.9),
TargetLynx™ and QuanOptimize™ (integrated in MassLynx version 4.2)
Thermo Fisher Scientific
(Waltham, MA, USA)
Xcalibur (version 4.3), BioPharma FinderTM (version 5.3) and PinpointTM (version 4.1)
Agilent Technologies
(Santa Clara, CA, USA)
MassHunter (version 12.0)
AB Sciex
(Framingham, MA, USA)
Analyst® (version 1.7.3), MultiQuantTM (version 3.0.3) and MRMPilotTM (version 2.1)
Biognosys AG
(Schlieren, DIE, ZH)
SpectronautTM (version 19), SpectromineTM (version 3), and SpectroDiveTM (version 12)
MacCoss Lab
(Seattle, WA, USA)
Skyline (version 24.1) 1
Cox Lab
(Martinsried, BY, GER)
MaxQuant (version 2.6.7.0) 1,2
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Döring, S.; Weller, M.G.; Reinders, Y.; Konthur, Z.; Jaeger, C. Challenges and Insights in Absolute Quantification of Recombinant Therapeutic Antibodies by Mass Spectrometry: An Introductory Review. Antibodies 2025, 14, 3. https://doi.org/10.3390/antib14010003

AMA Style

Döring S, Weller MG, Reinders Y, Konthur Z, Jaeger C. Challenges and Insights in Absolute Quantification of Recombinant Therapeutic Antibodies by Mass Spectrometry: An Introductory Review. Antibodies. 2025; 14(1):3. https://doi.org/10.3390/antib14010003

Chicago/Turabian Style

Döring, Sarah, Michael G. Weller, Yvonne Reinders, Zoltán Konthur, and Carsten Jaeger. 2025. "Challenges and Insights in Absolute Quantification of Recombinant Therapeutic Antibodies by Mass Spectrometry: An Introductory Review" Antibodies 14, no. 1: 3. https://doi.org/10.3390/antib14010003

APA Style

Döring, S., Weller, M. G., Reinders, Y., Konthur, Z., & Jaeger, C. (2025). Challenges and Insights in Absolute Quantification of Recombinant Therapeutic Antibodies by Mass Spectrometry: An Introductory Review. Antibodies, 14(1), 3. https://doi.org/10.3390/antib14010003

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop